CN111565801B - Peptide immunogens targeting membrane-bound IgE for the treatment of IgE-mediated allergic diseases and dosage forms thereof - Google Patents

Peptide immunogens targeting membrane-bound IgE for the treatment of IgE-mediated allergic diseases and dosage forms thereof Download PDF

Info

Publication number
CN111565801B
CN111565801B CN201780098083.4A CN201780098083A CN111565801B CN 111565801 B CN111565801 B CN 111565801B CN 201780098083 A CN201780098083 A CN 201780098083A CN 111565801 B CN111565801 B CN 111565801B
Authority
CN
China
Prior art keywords
ige
empd
seq
peptide
peptide immunogen
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN201780098083.4A
Other languages
Chinese (zh)
Other versions
CN111565801A (en
Inventor
王长怡
林峰
陈君伯
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
United Biomedical Inc
Original Assignee
United Biomedical Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by United Biomedical Inc filed Critical United Biomedical Inc
Publication of CN111565801A publication Critical patent/CN111565801A/en
Application granted granted Critical
Publication of CN111565801B publication Critical patent/CN111565801B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/35Allergens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0008Antigens related to auto-immune diseases; Preparations to induce self-tolerance
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/42Proteins; Polypeptides; Degradation products thereof; Derivatives thereof, e.g. albumin, gelatin or zein
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/42Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins
    • C07K16/4283Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins against an allotypic or isotypic determinant on Ig
    • C07K16/4291Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins against an allotypic or isotypic determinant on Ig against IgE
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55505Inorganic adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55561CpG containing adjuvants; Oligonucleotide containing adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55566Emulsions, e.g. Freund's adjuvant, MF59
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6031Proteins
    • A61K2039/6075Viral proteins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Microbiology (AREA)
  • Genetics & Genomics (AREA)
  • Mycology (AREA)
  • Engineering & Computer Science (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pulmonology (AREA)
  • Zoology (AREA)
  • Rheumatology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • Wood Science & Technology (AREA)
  • Inorganic Chemistry (AREA)
  • Toxicology (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Cell Biology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicinal Preparation (AREA)

Abstract

The present disclosure relates to IgE EMPD peptide immunogen constructs and dosage forms thereof for use in the treatment of IgE-mediated allergic diseases. IgE EMPD peptide immunogen constructs have B cell epitope peptides of more than 20 amino acids, preferably cyclic, linked via an optional spacer to a heterologous T helper cell (Th) epitope derived from a pathogen protein. The peptide immunogen constructs and dosage forms thereof are capable of stimulating the production of highly specific antibodies in the vaccinated host against IgE EMPD peptides and cross-react with membrane-bound IgE on B lymphocytes that differentiate committed to IgE secretion. The peptide immunogen constructs and dosage forms thereof induce antibody apoptosis of IgE-expressing B cells in the vaccinated host and mediate antibody-dependent cellular cytotoxicity (ADCC), resulting in reduced levels of specific IgE and total IgE of the vaccinated host antibodies for effective treatment of IgE-mediated allergic pathologies.

Description

Peptide immunogens targeting membrane-bound IgE for the treatment of IgE-mediated allergic diseases and dosage forms thereof
Technical Field
The present disclosure relates to peptide immunogen constructs targeting the extracellular membrane proximal domain of membrane-bound IgE (or IgE EMPD) and dosage forms thereof as a universal vaccine for the treatment and/or prevention of IgE-mediated allergic diseases.
Background
Allergy, also known as immunoglobulin E (IgE) mediated allergic disease, is a number of diseases caused by the high sensitivity of the immune system to certain substances in the environment, often causing little or no problems in most people. These diseases include allergies to drugs, foods and insects, allergic rhinitis (hay fever), atopic dermatitis, allergic asthma, conjunctivitis, eczema, urticaria (urticaria) and acute allergies (website: en. Wikipedia org/wiki/Allergy). Many symptoms are often due to allergies and may include redness of the eyes, itchy rash, sneezing, running nose, shortness of breath, or swelling.
The prevalence of allergic diseases is increasing. Allergies were considered a rare disease in the beginning of the 20 th century. However, since then, there have been several factors that have triggered a dramatic rise in the prevalence of allergic diseases. The respiratory tract characterization is most common, affecting up to 30% of the general population. According to World Health Organization (WHO) statistics, hundreds of millions of people in the world suffer from rhinitis and it is estimated that 2.35 hundred million people suffer from asthma (website: www.who.int/media/pictures/fs 307/en/index. Html). The social costs of allergic diseases are considerable, mainly due to the high prevalence of allergic rhinoconjunctivitis and the productivity losses associated therewith. Swedish studies estimated that productivity loss due to rhinitis is up to 27 million Euro annually in Swedish alone, while U.S. studies demonstrated that the loss to the United states employer in disease was greatest with rhinitis (Larsen JN et al 2016).
Allergic reactions are abnormally severe immune reactions where the immune system is protected against the threat of antigens (allergens) that were originally harmless to humans (website: en. Wikipedia. Org/wiki/Allergen). Specifically, an allergen is an antigen capable of stimulating a type I allergic response in an atopic individual through the IgE response. Allergens may come from a variety of sources (e.g., dust mite feces, pollen, pet dander, certain foods, or chemical/physical irritants). Food allergy is not as common as food sensitivity, but certain foods (e.g., peanuts (beans), nuts, seafood, and shellfish) are responsible for severe allergy in many people.
Allergy is a systemic immune disorder caused by the activation of an adaptive immune response against common allergens. IgE plays an important role in mediating the type I allergic reactions responsible for the induction of IgE-mediated allergic diseases. IgE-mediated allergic diseases are characterized by the presence of allergen-specific IgE antibodies and eosinophilic inflammation. The allergic reaction is biphasic, an immediate type reaction occurs within minutes after allergen exposure, and a late reaction occurs after hours. Immediate type reactions are caused by ready-made mediators (e.g., histamine, proteases, chemokines, heparin) released by basophils and mast cells upon cross-linking of IgE bound to high affinity receptors located on the cell surface. Late allergic reactions are caused by mobilization and aspiration of inflammatory cells (e.g., eosinophils, basophils, neutrophils, and monocytes).
Allergens cause an increase in serum total free immunoglobulin E (IgE) and allergen-specific IgE levels in allergy-prone individuals. Allergen-specific IgE-mediated type I allergic reactions are important for the pathogenic mechanism of IgE-mediated allergic diseases (fig. 1). IgE sensitizes mast cells and basophils by binding to high affinity IgE receptors (fceri) located on the surface of those effector cells. The antigen binds to IgE that has bound to fceri on mast cells, resulting in cross-linking of the bound IgE, and aggregation of fceri below. The crosslinked receptors initiate a signaling cascade with rapid degranulation. Mast cells and basophils release stored histamine and then synthesize and release bradykinin, prostaglandins, leukotrienes, cytokines and other mediators of inflammation. They further attract and activate inflammatory cells that produce allergic symptoms and up regulate IgE synthesis by B cells, thereby promoting exacerbation of sensitivity. IgE-fceri interactions and degranulation are important for the development of type I allergic reactions and atopic asthma.
Like other immunoglobulin isotypes, igE exists in 2 forms (secreted serum immunoglobulin form and membrane bound form (mIgE)). Studies of gene fragments encoding membrane-anchored peptides of mouse and human mIgEs have shown that the difference between mIgE and secreted IgE is that the mIgE comprises three additional regions: (1) A central conserved stretch of 25 hydrophobic, uncharged amino acid residues that spans the cell membrane; (2) a cytoplasmic tail at the carboxy terminus; and (3) the amino-terminal extracellular portion of an mIgE membrane anchor fragment. In humans, epsilon chains of mIgE located on the surface of B lymphocytes exist in short and long isoforms. The short isoform contains 15 amino acids located in the proximal domain of the outer membrane of the mIgE cell, called IgE EMPD, while the long isoform contains an additional fragment of 52 amino acid residues, totaling 67 amino acids in EMPD. These two isoforms are the result of alternative splicing between a donor site located 3' to the CH4 exon and two acceptor sites (156 bp apart and located about 2000 nucleotides downstream of the CH4 exon). Detection of IgE-producing myeloma and tonsil B cells treated with IL-4 plus CD40 showed that long form transcripts were 100-fold higher than shorter form transcripts (Peng et al 1992and Zhang et al 1992); while no short forms were detected at the protein level (Peng et al, 1992). IgE EMPD is specific for the mIgE form, which was not found in secreted serum IgE (fig. 2).
Current clinical guidelines for IgE-mediated allergic disease treatment include combinations of patient education, allergen prevention, drug therapy, allergen-based immunotherapy, and therapeutic targets for IgE, but these therapeutic options have their limitations. For example, although allergen prevention is actually indicated as much as possible, adequate symptom control is still difficult to achieve with allergen prevention alone. Furthermore, even though safe and inexpensive drugs are available for treating allergic symptoms, many patients report that the use of these drugs is not sufficient to control the symptoms. Importantly, drug treatment has no effect on disease progression and repeated administrations of treatment are necessary as long as symptoms are prevalent, often implying lifelong treatment. Only typical immunogen-based immunotherapy has the potential to ameliorate disease and is considered the most desirable therapeutic strategy.
Allergen-based immunotherapy (AIT) involves the incremental subcutaneous injection of increased doses of allergen in order to suppress symptoms caused by subsequent re-exposure to such allergen. Under natural exposure conditions, the allergen content of the immune system present in the mucosa is relatively low, but it can be effective to stimulate allergic reactions and develop allergic symptoms within minutes. In contrast, when an allergen is administered as an immunotherapy, the amount of allergen is relatively high, wherein one dose administered in the immunotherapy corresponds to about 100 times the estimated maximum annual intake by natural exposure. Combining the quantitative differences with different pathways into the human body, profound effects are exhibited on the immune system, which reacts by eliciting immune tolerance to allergens.
The original administration form of AIT is by subcutaneous injection, and this treatment regimen has traditionally been carried out in two stages: (1) An initial incremental administration phase and (2) a subsequent maintenance phase. The incremental dosing phase is an individual adjustment of the dose where increasing doses are administered for the purpose of slowly establishing tolerance and carefully assessing the sensitivity of the patient. The maximum tolerated dose or the recommended highest dose is administered first, followed by the dose throughout the maintenance phase.
Two mechanisms are believed to play a major role in AIT: (1) immune bias and (2) induction of regulatory T cells. The relative contributions of immune bias and regulatory T cells have not been determined, but the end result is a reduction and in some cases even elimination of the ability to cause allergic reactions in the response to allergen exposure.
Immune bias is a term used to represent a modified immune response to allergen exposure in which allergen-specific T helper I (Th 1) cells are mobilized and stimulated but sacrificed for Th2 cells. Th1 cells produce interferon gamma (IFN-gamma), which stimulates B cells to produce IgG instead of IgE, and IgG is unable to elicit an allergic response.
Regulatory T cells are a diverse group of T cells that are active in the regulation of immune responses. An increase in allergen-specific cd4+cd25+ regulatory T cells in peripheral blood after AIT has been demonstrated. These cells produce Interleukin (IL) -10 and Transforming Growth Factor (TGF) - β and have the potential to suppress local Th2 cell responses and redirect antibody class switching, biasing IgG4 (IL 10 isotype switching factor) and IgA (TGF- β isotype switching factor). Allergen-specific IgG4 antibodies block allergen presentation to Th2 cells and additionally block allergen-induced activation of mast cells and basophils, thereby significantly attenuating the allergic response.
Although antigen-based immunotherapy may be effective, there are still serious problems and unmet needs associated with AIT of IgE-mediated allergic diseases. First, all injections of AIT are performed in the clinic, as there is a slight risk of eliciting allergic reactions, which may become serious or even life threatening if not timely and properly treated. Second, only a few allergen products have been clinically proven to be disease ameliorated by AIT. Third, only a few specific allergen structures have been described and the definition of an allergen is based mainly on functional criteria capable of eliciting IgE responses in susceptible individuals. Thus, allergens are generally defined by the immune system of an individual patient, and therefore, even if most allergic patients have IgE antibodies specific to a relatively limited number of major allergens, any immunogenic protein (antigen) has the potential to elicit an allergy. Fourth, each patient has a unique pattern of sensitization to allergen molecules and epitopes. Fifth, all commercial allergen products are manufactured using aqueous extraction of allergy inducing substances derived from natural sources (e.g., pollen, house dust mite cultures, animal hair and/or dander, or insect venom), and such natural sources are intrinsically variable in composition. Thus, allergen products for AIT are not universal and have variability in their composition, igE binding capacity and quality control management among manufacturers. There is no international standard in effect. This means that products from different manufacturers may differ in effect in patients, so that clinical outcome cannot be inferred directly from one allergen product to another.
Recent studies review allergen-based immunotherapy: future treatments for allergy (Drug Discovery Today Volume 21,Issue 1,January2016,Pages 26-37) are included herein by reference. Wherein all supporting files can be found in the statements made in this background section.
In addition to AIT, therapeutic targets for IgE molecules have been investigated in the treatment of IgE-mediated allergic diseases.
Therapeutic targets utilizing serum soluble IgE of anti-IgE monoclonal antibodies have been shown to be effective in IgE-mediated allergic disease treatment. Currently, omalizumab(a recombinant humanized monoclonal antibody) has been approved for the treatment of moderate to severe persistence in adults and young peopleAllergic asthma. Omalizumab prevents the allergic cascade by binding to circulating unbound free IgE and prevents it from binding to IgE fceri on the surface of immune effector cells. Treatment with Omalizumab results in a significant decrease in free IgE levels and down-regulation of the IgE receptors of cells (Chang et al, 2007). Although treatment with Omalizumab has proven effective, there are also limitations. Specifically, omalizumab neutralizes free IgE in serum, but does not affect IgE production. Therefore, omalizumab must be administered frequently and chronically to maintain adequate serum IgE inhibition.
Therapeutic targets for the extracellular membrane proximal domain (IgE EMPD) of membrane-bound IgE have also been studied to treat IgE-mediated allergic diseases. B Cell Receptor (BCR) cross-linking in the absence of additional co-stimulatory signals can lead to B cell apoptosis. For immature B cells, B cell apoptosis depletion by BCR cross-linking has been widely described as a mechanism by which autoreactive B cells are removed from B cell libraries. anti-IgE EMPD monoclonal antibodies, such as 47H4 (bright et al 2010), 4B12, and 26H2 (Chen et al 2010), have been shown to cross-link IgE BCR and cause B-cell apoptosis that expresses mIgE (fig. 2). Brightbill et al also found that therapeutic delivery of 47H4 in vivo reduced established IgE responses, as observed in the Nocardia infection and allergic asthma model of Brazil (Chen et al 2010). In order to deplete IgE lineage B cells to reduce serum IgE, several IgE-EMPD-targeting antibodies and epitopes have been studied and identified, especially over a long form region of additional 52 amino acids (Chen et al 2010, chang et al 2015, chen et al 2002). One team indicated that the use of hepatitis B virus core antigen (HBcAg) as a vector carried IgE EMPD fragments as inserts to induce specific IgE EMPD antibody responses in BALB/c mice. This cloning construct spontaneously assembles into Viroid Particles (VPLs) that present various IgE EMPD fragments on top of the spike protein (spike) of the VPLs to gain immunogenicity. IgG antibodies purified from serum of immunized mice can be caused to express mIgE. Fc via the BCR-dependent apoptosis protease pathway by using purified mouse spleen NK cells as effector cells L Apoptosis of Ramos cells in the expression of mige. Fc L Is responsible for Antibody Dependent Cellular Cytotoxicity (ADCC) (Lin, et al 2012). The above-described methods have attracted some interest in the development of allergy therapeutic vaccines. However, antigen expression systems are inefficient, producing antibodies to most targeting vector VLPs, and antigen and delivery systems are far from suitable for development of effective vaccines for industrial and clinical use.
In view of the above, there is a need to develop immunotherapeutic approaches to treat and/or prevent IgE-mediated allergic diseases, which are allergen-independent, capable of eliciting highly specific immune responses against IgE, easy to administer to patients, capable of being manufactured according to strict Good Manufacturing Practice (GMP), and cost-effective in worldwide applications, instead of practicing AIT for centuries.
Reference is made to:
1.LARSEN,J.N.,et al.“Allergy Immunotherapy:The Future of Allergy Treatment”,Drug Discovery Today,1:26-37(2016).
2.PENG,C.,et al.“A New Isoform of Human Membrane-Bound IgE”,J.Immunol.148:129-136(1992).
3.ZHANG,K.,et al.“Two unusual forms of human immunoglobulin E encoded by alternative RNA splicing of epsilon heavy chain membrane exons”,J.Exp.Med.,176:233-243(1992).
4.CHEN,J.B.,et al.“Unique epitopes on CεmX in IgE-B cell receptors are potentially applicable for targeting IgE-committed B cells”,J.Immunol,184:1748-1756(2010).
5.LIN,C.J.,et al.“Cεm X peptide-carrying HBcAg virus-like particles induced antibodies that down-regulate mIgE-B lymphocytes”,Mol.Immunol.,52:190-199(2012).
6.CHANG,T.W.,et al.“C(Epsilon)m X Peptides for Inducing Immune Responses to Human mIgE on B Lymphocytes”,US Patent No.8,974,794 B2(2015).
7.CHEN,H.Y.,et al.“Monoclonal Antibodies against the CεmX Domain of Human Membrane-Bound IgE and Their Potential Use for Targeting IgE-Expressing B Cells”,Immunol.,128:315-324.(2002).
8.BRIGHTBILL,H.D.,et al.“Antibodies specific for a segment of human membrane IgE deplete IgE-producing B cells in humanized mice”,J Clin.Invest.,120:2218-2229(2010).
9.LU,C.S.,et al.“Generating allergen-specific human IgEs for immunoassays by employing humanεgene knockin mice”,Allergy,70:384-390(2015).
10.WU,P.C.,et al.“The IgE gene in primates exhibits extraordinary evolutionary diversity”,Immunogenetics,64:279-287(2012).
11.CHANG,T.W.,et al.“Anti-IgE Antibodies for the Treatment of IgE-Mediated Allergic Diseases”,Advances in Immunology,93:63-119(2007).
12.TRAGGIAI,E.,et al.“An efficient method to make human monoclonal antibodies from memory Bcells:potent neutralization of SARS coronavirus”,Nature Medicine,10:871-875(2004).
13.“Asthma Fact Sheet”World Health Organization website,website address:www.who.int/mediacentre/factsheets/fs307/en/index.html(accessed August 18,2017).
14.“2016Appen dix to GINA Report”Global Initiative For Asthma website,website address:ginasthma.org/wp-content/uploads/2016/04/GINA-Appendix-2016-final.pdf(accessed August 18,2017).
15.“Allergy”Wikipedia,The Free Encyclopedia,website address:en.wikipedia.org/wiki/Allergy(accessed August18,2017).
16.website:en.wikipedia.org/wiki/Allergen)
summary of The Invention
The present disclosure relates to individual peptide immunogen constructs targeting the extracellular membrane proximal domain (IgE EMPD) of membrane-bound IgE for the treatment and/or prevention of IgE-mediated allergic diseases. The disclosure also relates to compositions comprising such peptide immunogen constructs, methods of making and using such peptide immunogen constructs, and antibodies produced using such peptide immunogen constructs.
The disclosed peptide immunogen constructs comprise about 20 or more amino acids. The peptide immunogen construct comprises a B cell epitope of 67 amino acid sequences from full length IgE EMPD (SEQ ID NO: 1). This B cell epitope may be linked through an optional heterologous spacer to a heterologous T helper cell (Th) epitope derived from a pathogen protein. The disclosed peptide immunogen constructs may stimulate the production of highly specific antibodies against IgE EMPD, and such antibodies may bind to recombinant IgE EMPD-containing proteins, igE EMPD on B cells with mIgE, and/or recombinant soluble IgE EMPD proteins comprising the Fc portion of human IgG1, and IgE EMPD of human membrane-bound IgE (referred to as "γ1-em 67"). The disclosed peptide immunogen constructs are useful as a cost-effective, general-purpose immunotherapy for global patients with IgE-mediated allergic diseases, independent of the allergen.
The B cell epitope portion of the peptide immunogen construct has an amino acid sequence derived from the full-length IgE EMPD sequence (SEQ ID NO: 1). In some embodiments, the B cell epitope has a sequence comprising an internal intramolecular loop formed by endogenous cysteines (C18-C39) according to the numbering of the full length IgE EMPD sequence (SEQ ID NO: 1). In certain embodiments, the B cell epitope has the amino acid sequence of IgE EMPD-1-39 (SEQ ID NO: 5), igE EMPD-7-40 (SEQ ID NO: 6), igE EMPD-19-38 (SEQ ID NO: 8), or IgE EMPD-1-40 (SEQ ID NO: 9).
The peptide immunogen constructs of the present disclosure may contain heterologous Th epitope amino acid sequences derived from pathogen proteins (e.g., SEQ ID NOs:59 to 87). In certain embodiments, the heterologous Th epitope is derived from a pathogen in nature, for example: diphtheria toxin (SEQ ID NO: 63), plasmodium falciparum (SEQ ID NO: 64), cholera toxin (SEQ ID NO: 66). In other embodiments, the heterologous Th epitope is an idealized artificial Th epitope derived from measles virus fusion proteins (MVF 1 to 5) or hepatitis B surface antigen (HBsAg 1 to 3) in the form of a single sequence or a combined sequence (e.g., SEQ ID NOS: 70, 69 and 71).
In some embodiments, the peptide immunogen construct comprises a B cell epitope from IgE EMPD linked to a heterologous T helper cell (Th) epitope through an optional heterologous spacer. In certain embodiments, the peptide immunogen construct comprises a B cell epitope from IgE EMPD-1-40 (SEQ ID NO: 9) having more than about 20 amino acids linked by an optional heterologous spacer to a heterologous Th epitope derived from a pathogen protein (e.g., SEQ ID NOs:59 to 87). In some embodiments, the optional heterologous spacer is a molecule or chemical structure capable of linking two amino acids and/or peptides together. In certain embodiments, the spacer is a naturally occurring amino acid, a non-naturally occurring amino acid, or a combination thereof. In a specific embodiment, the peptide immunogen construct has the sequence of SEQ ID NO:88-95, 98-124 and 130.
The disclosure also relates to compositions comprising IgE EMPD peptide immunogen constructs. In some embodiments, the disclosed compositions comprise more than one IgE EMPD peptide immunogen construct. In certain embodiments, the compositions comprise a mixture of IgE EMPD G1-C39 peptide immunogen constructs (e.g., any combination of SEQ ID NOS: 88-95, 98-124 and 130) to encompass a broad genetic background for the patient. Compositions comprising a mixture of peptide immunogen constructs may result in a higher percentage of response after vaccination compared to compositions comprising only a single peptide immunogen construct to treat IgE-mediated allergic diseases.
The present disclosure also relates to pharmaceutical compositions, including vaccine dosage forms, for the treatment and/or prevention of IgE-mediated allergic diseases. In some embodiments, the pharmaceutical composition comprises the disclosed peptide immunogen constructs in the form of a stabilized immunostimulatory complex formed by mixing a CpG oligomer and a composition comprising the peptide immunogen complex to form by electrostatic binding. Such stabilized immunostimulatory complexes may further enhance the immunogenicity of the peptide immunogen construct. In some embodiments, the pharmaceutical composition comprises an adjuvant, such as a mineral salt, including alum gel (ALHYDROGEL), aluminum phosphate (ADJUPHOS), or a water-in-oil emulsion including MONTANIDEISA 51 or 720.
The disclosure also relates to antibodies directed against the disclosed IgE EMPD peptide immunogen constructs. In particular, when administered to a subject, the peptide immunogen constructs of the present disclosure may stimulate the production of highly specific antibodies that cross-react with IgE EMPD-1-52 amino acid sequences (SEQ ID NO: 2), igE EMPD 1-67 amino acid sequences (SEQ ID NO: 1), and fragments thereof (e.g., SEQ ID NO:5 and 6). Highly specific antibodies made using the peptide immunogen constructs may cross-react with recombinant IgE EMPD-containing proteins, γ1-em67, and/or IgE EMPD on B cells with membrane-bound IgE. The disclosed antibodies utilize high specificity binding to IgE EMPD, with little, if any, targeting of heterologous Th epitopes for immunogenicity enhancement, in stark contrast to antibodies made using conventional proteins or other biological carriers for peptide antigenicity enhancement.
The present disclosure also includes methods of using the disclosed peptide immunogen constructs and/or antibodies directed against the peptide immunogen constructs to treat and/or prevent IgE-mediated allergic diseases. In some embodiments, a method for treating and/or preventing IgE-mediated allergic diseases comprises administering to a host a composition comprising the disclosed peptide immunogen constructs. In certain embodiments, the compositions used in the methods comprise the disclosed peptide immunogen constructs in the form of stabilized immunostimulatory complexes formed by electrostatic binding using negatively charged oligonucleotides (e.g., cpG oligomers), which may be complexed with further supplemental adjuvants (optionally mineral salts or oils as adjuvants) for administration to IgE-mediated allergic disease patients. The disclosed methods also include administration of the peptide immunogen constructs to a dosage regimen, dosage form and route of administration to a host at risk of IgE-mediated allergic disease or that has developed.
Drawings
FIG. 1 is a diagram depicting the mechanism of the IgE mediated allergic disease pathway. The initial mature B cells begin to express membrane-bound IgE (mIgE). Upon encountering an allergen, these cells become activated with the aid of homologous T Helper (TH) cells, which provide the B cells with the necessary costimulatory signals and cytokines. Activated allergen-specific B cells are transformed into IgE-targeted B cells (IgE-committed B cells) expressing mIgE by class switching recombination (class-switching recombination) with the aid of a variety of cytokines, such as IL-4 and IL-13. Those IgE direct B cells to eventually differentiate into IgE-secreting plasma cells. Most IgE secreting plasma cells are short lived and die after moving to the site of inflammation; however, some of the longevity cells migrate to the corresponding microenvironment in the bone marrow. Allergen-specific IgE secreted from plasma cells binds to high-affinity ige.fc receptors (fceri) located on the surface of blood basophils and tissue mast cells. Aggregation of IgE bound to FcgRI by allergens stimulates basophil or mast cell degranulation and releases vehicles (e.g., histamine, leukotrienes, PGD 2, tryptase and various cytokines) which trigger immediate allergic reactions and promote mobilization of various cell types (e.g., TH2 cells and eosinophils).
Fig. 2A and 2B are diagrams showing structural differences between secreted IgE and membrane-bound IgE (mIgE) and the rationale for depletion of mIgE B cells by targeting IgE EMPD. Figure 2A shows IgE expressed in two forms: secreted IgE and membrane-bound IgE (mIgE). Secreted IgE is captured on the cell surface of basophils and mast cells by fceri, whereas mIgE is present only on IgE-targeted B cells as part of B cell receptors. The outer membrane proximal domain (EMPD) of mIgE is a 67 amino acid peptide fragment (SEQ ID NO: 1) located between the CH4 domain and the transmembrane region found on mIgE B cells only. The underlined amino acids represent residues found in the short isoforms of EMPD. The uniqueness of IgE EMPD provides an attractive site for targeting mIgE and mIgE B cells. Figure 2B shows the mechanism of depletion of mIgE B cells by targeting IgE EMPD, which results in inhibition of allergen-specific IgE production before the mIgE B cells differentiate into new IgE-secreting plasma cells. Existing IgE-secreting plasma cells with limited life will eventually die out successively, resulting in a gradual decrease in total IgE and allergen-specific IgE.
Fig. 3 is a flow chart identifying the development of vaccine dosage forms from discovery to commercialization (industrialization) according to certain embodiments disclosed herein. As summarized in this figure, the present disclosure includes peptide immunogen design, peptide composition design, vaccine dosage form design, in vitro functional antigenicity study design, in vivo immunogenicity and efficacy study design, and clinical trial planning design. The detailed evaluation and analysis of each step led to a series of experiments leading to the commercialization of safe and effective vaccine dosage forms.
FIG. 4 is a graph illustrating the kinetics of antibody responses over a period of 8 weeks in guinea pigs immunized with different IgE EMPD peptide immunogen constructs (SEQ ID NOS: 88 to 94, 96 and 97). Serum was diluted from 1:100 to 1:100000 with 10-fold serial dilutions. ELISA plates were coated with IgE EMPD 1-39 peptide (SEQ ID NO: 5) in an amount of 0.5. Mu.g peptide per well. By A 450 A with a threshold of 0.5 450 The titer of the test serum was calculated by linear regression analysis of (C) in Log 10 And (3) representing.
FIG. 5 is a graph illustrating titration curves for various purified polyclonal anti-IgE EMPD antibodies generated using different IgE EMPD immunogen constructs (SEQ ID NOS: 88 to 94, 96 and 97). ELISA plates were coated with recombinant IgE EMPD (SEQ ID NO: 1) containing protein, gamma 1-em67 (SEQ ID NO: 1). Polyclonal anti-IgE EMPD antibodies purified from guinea pig serum by protein A chromatography were diluted from 100. Mu.g/mL to 0.0238ng/mL using 4-fold serial dilutions. EC calculation of each polyclonal anti-IgE EMPD antibody using nonlinear regression of 4-parameter logistic curve fit 50 In the immunogen construct SEQ ID No:89 and 93 show the best binding efficiency.
FIGS. 6A through 6C contain purified polyclonal antibodies from pooled guinea pig sera from animals immunized with IgE EMPD immunogen constructs to express mIgE. Fc L (left) or mIgE. Fc s Flow cell histogram of the binding of Ramos cell line cells (flow cytometry histograms) on the right. Polyclonal anti-IgE EMPD antibodies purified from guinea pig serum using protein A chromatography were used at a concentration of 10 μg/mL compared to IgE EMPD B cell epitope peptides having a size of less than 20 amino acid residuesImmunogen constructs 96 and 97, immunogen constructs with SEQ ID NO from 88 to 94 showed a high activity against mIgE. Fc L B cells bind quite. Fig. 6A contains the sequence from the anti-IgE EMPD immunogen construct SEQ ID NO:88 to 90. Fig. 6B comprises the sequence derived from the immunogenic construct SEQ ID NO: bar graphs of polyclonal antibodies 91 to 93. Fig. 6C contains the sequence from the anti-IgE EMPD immunogen construct SEQ ID NO: 94. bar graphs of polyclonal antibodies at 96 and 97.
FIG. 7 is a schematic diagram showing the elicitation of mIgE.Fc expression using various polyclonal anti-IgE EMPD antibodies against IgE EMPD immunogen constructs (SEQ ID NOS: 88 to 93) L Apoptosis pattern of Ramos cells of (a). By% Annexin V + /PI - Indicating the level of apoptosis. Polyclonal anti-IgE EMPD antibodies purified from guinea pig serum by protein a chromatography were diluted from 1000 to 62.5ng/mL using 2-fold serial dilutions. Use of humanized anti-IgE monoclonal antibodies As a positive control. EC of polyclonal anti-IgE EMPD antibodies per group 50 Shown below the figure, calculated using nonlinear regression of 4-parameter logic Luo Ji t-curve fit, shown to elicit mige. Fc with the immunogenic construct of SEQ ID NOs with 88, 90 and 93 sequence identification numbers L Optimal efficacy of B cell apoptosis.
FIG. 8 is a graph showing the elicitation of mIgE.Fc expression by polyclonal anti-IgE EMPD antibodies against IgE EMPD immunogenic constructs (SEQ ID NOS: 88 to 93) at an effector cell/target cell ratio of 1/30 L Antibody Dependent Cellular Cytotoxicity (ADCC) histogram of Ramos cells. Polyclonal anti-IgE EMPD antibodies purified from guinea pig serum by protein A chromatography were used at a concentration of 10. Mu.g/mL. IL-2 stimulated mouse spleen cells were used as effector cells, while the mouse anti-IgE monoclonal antibody 5D5 was used as a positive control.
FIG. 9 is a diagram illustrating epitope identification for fine specificity analysis using immune serum from guinea pigs using overlapping 10-mer peptides covering amino acids 9-50 of IgE EMPD in ELISA. The primary epitope recognized by antibodies in serum samples is specific for the cyclic structural region representing IgE EMPD. This figure illustrates the internal cyclic structure formed by intramolecular disulfide bonds between amino acids C18 and C39 in native IgE EMPD. Serum was diluted 1:1000 for epitope identification. ELISA plates were coated with 10-mer peptide (0.5. Mu.g peptide per well). By using the sequence of the sequence having SEQ ID NO from the previous use: 88. immune serum samples collected from 89, 93, 96 and 97 immunized guinea pigs were subjected to epitope identification studies to identify reaction sites and labeled accordingly.
Fig. 10 is a diagram illustrating an experimental design for assessing papain-induced primary and secondary immune responses following immunization with the peptide immunogen constructs of the present disclosure. Human IGHE knock-in hybrid mice were vaccinated three times intramuscularly (im) with IgE EMPD peptide immunogen constructs at weeks 0, 3 and 5. In the primary IgE response model, mice were challenged subcutaneously (sc) with papain/TiterMax Gold at week 10 and papain-specific human IgE (hIgE) was determined at week 12 (as shown in fig. 13). In the secondary IgE response, mice were challenged subcutaneously with papain/TiterMax Gold again at week 16, and papain-specific human IgE (hIgE) was determined at week 18 (as shown in fig. 14). Serum from mice receiving immunization was also tested for anti-IgE antibody production (fig. 11) and serum IgE change (fig. 12) throughout the study.
FIG. 11 is a graph illustrating the kinetics of anti-IgE antibody production over a 20 week period using the experimental design described in FIG. 10. Specifically, this graph shows the antibody response in human IGHE knock-in mice (hIGHE x Balb/c, each group n=8) which was immunized three times intramuscularly with IgE EMPD immunogen construct (SEQ ID NO:88 or 93) at the indicated dose (100 μl per immunization) at weeks 0, 3 and 5 and challenged subcutaneously with papain/TiterMax at weeks 10 and 16. Mouse serum was diluted from 1:100 to 1: (4.19X10) with 4-fold serial dilution 8 ). ELISA plates were coated with recombinant IgE EMPD-containing protein gamma 1-em 67. Benefit (benefit)Non-linear regression fitted with 4-parameter logistic curves was used to calculate the titers of test serum as a function of Log (EC 50 ) And (3) representing.
FIG. 12 is a graph illustrating the change in serum IgE over a 20 week period using the experimental design described in FIG. 10. Specifically, this graph shows serum IgE levels in human IGHE knock-in mice (hIGHE x Balb/c, each group n=8) which were vaccinated three times intramuscularly with IgE EMPD immunogen constructs (SEQ ID NO:88 or 93) at the indicated doses (100 μl per vaccination) at weeks 0, 3 and 5 and challenged subcutaneously with papain/TiterMax at weeks 10 and 16. Serum IgE was assayed in a human IgE (hIgE) quantitative ELISA. Mouse serum was diluted 1:20. Standard curves for hIgE production purified from U266 myeloma cells were used. By mixing A with 450 IgE concentration was calculated by interpolation into a standard curve generated by nonlinear regression using a 4-parameter logistic curve fit.
FIG. 13 is a graph showing inhibition of papain-specific human IgE (hIgE) production in a primary IgE reaction determined at week 12 using the experimental design described in FIG. 10. Specifically, human IGHE knock-in hybrid mice (hIGHE x Balb/c, each group n=8) were immunized intramuscularly with IgE EMPD immunogen constructs (SEQ ID NO:88 or 93) three times at the indicated doses (100. Mu.L per immunization) at weeks 0, 3 and 5. Serum papain-specific hIgE was assayed in a quantitative ELISA. Mouse serum was diluted 1:10. Standard curves were generated using monoclonal chimeric papain-specific hIgE. By mixing A with 450 The papain-specific hIgE concentration was calculated by interpolation into a standard curve generated by nonlinear regression using a 4-parameter logistic curve fit.
FIG. 14 is a graph showing inhibition of papain-specific human IgE (hIgE) production in a secondary IgE response determined at week 18 using the experimental design described in FIG. 10. Specifically, human IGHE knock-in hybrid mice (hIGHE x Balb/c, each group n=8) were immunized intramuscularly with IgE EMPD immunogen constructs (SEQ ID NO:88 or 93) three times at the indicated doses (100. Mu.L per immunization) at weeks 0, 3 and 5. Determination of serum in quantitative ELISAPapain-specific hIgE. Mouse serum was diluted 1:10. Standard curves were generated using monoclonal chimeric papain-specific hIgE. By mixing A with 450 The papain-specific hIgE concentration was calculated by interpolation into a standard curve generated by nonlinear regression using a 4-parameter logistic curve fit.
Fig. 15 is a schematic diagram illustrating an experimental design to evaluate papain-induced sensitization and recall immune responses following immunization with the peptide immunogen constructs of the present disclosure. Human IGHE knockin mice were sensitized with papain/TiterMax Gold subcutaneous (sc) at week 0 and then three intramuscular immunizations with IgE EMPD peptide immunogen constructs were performed at weeks 3, 6 and 8. Papain-specific immune recall responses were elicited by intradermal footpad injection at week 12 using papain in PBS solution. Total IgE and papain-specific IgE/IgG levels were assessed between weeks 0 to 6 of the experiment (fig. 16), and papain-specific IgE levels were assessed at weeks 12, 13 and 14 (fig. 17).
Fig. 16 contains graphs showing the results of the papain sensitization of all human IGHE knockin hybrid mice using the experimental design described in fig. 15. Papain-specific mouse IgG titers were measured as Log (EC 50 ) Human IgE (ng/mL) was expressed and detected over a 6 week period. In addition, total IgE levels (ng/mL) increased due to bystander activation (bystander activation).
FIG. 17 is a graph showing inhibition of papain-specific human IgE (hIgE) production measured at weeks 12, 13 and 14 using the experimental design described in FIG. 15. Specifically, the figures show that the three times on weeks 3, 6 and 8 utilized a dose of 400 μg/mL (100 μl per immunization) of SEQ ID NO:88 or 93, sensitized human IGHE knockin mice (hIGHE x Balb/c, each group n=8). Serum papain-specific hIgE was assayed in a quantitative ELISA. Mouse serum was diluted 1:10. Standard curves were generated using monoclonal chimeric papain specific hIgE (ng/mL). By mixing A with 450 Interpolation to logical curve fitting with 4-parametersPapain-specific hIgE concentrations were calculated from standard curves generated by nonlinear regression.
Figures 18A and 18B are graphs showing immunogenicity of original immunotherapeutic allergy vaccine formulations in cynomolgus monkeys immunized with four doses of IgE EMPD immunogen construct (SEQ ID NO: 88) plus placebo control formulation at the indicated doses at weeks 0, 3 and 6 at 30, 100, 300 and 1000 μg per dose and evaluating anti-IgE EMPD titers by ELISA. FIG. 18A shows the use of a composition containing Montanide TM Antibody response in cynomolgus monkeys immunized with formulations of ISA 51 and CpG ODN. Fig. 18B shows antibody responses in cynomolgus monkeys immunized with dosage forms containing ADJUPHOS and CpG ODN.
FIG. 19 is a graph illustrating the kinetics of antibody responses over a period of 20 weeks in cynomolgus monkeys (2 males and 2 females per group) immunized 3 times intramuscularly with an immunogen construct (SEQ ID NO:125 or 126) at a dose of 300 μg/mL (500 μl per immunization) at weeks 0, 3 and 6. Macaca fascicularis serum was diluted from 1:100 to 1: (4.19X10) with 4-fold serial dilution 8 ). Using SEQ ID NO:5 coating ELISA plates. Non-linear regression calculation of test serum titers using 4-parameter logistic curve fitting to dilute the Log of factors 10 And (3) representing. The threshold was set at the average A of all serum samples diluted 1:100 450 Is 2 times as large as the above.
FIG. 20 is a graph illustrating the kinetics of IgG, igA and IgM antibody responses over a 20 week period in cynomolgus monkeys (2 males and 2 females per group) immunized 3 times intramuscularly with IgE EMPD immunogen constructs (SEQ ID NO:125 or 126) at doses of 300 μg/mL (500 μl per immunization) at weeks 0, 3 and 6. Macaca fascicularis serum was diluted from 1:100 to 1: (4.19X10) with 4-fold serial dilution 8 ). ELISA plates were coated with IgE EMPD 1-39 peptide (SEQ ID NO: 5). Calculating titers by interpolating threshold values into 4-parameter logistic curves generated from each test serum data, expressed as Log 10 And (3) representing. The threshold was set at the average A of all serum samples diluted 1:100 450 Is 2 times as large as the above.
FIG. 21 is a graph illustrating the use of 300 μg/mL at weeks 0, 3 and 6 (500 μ per immunization)L) IgE EMPD immunogen construct (SEQ ID NO:125 or 126) patterns of serum IgE levels change over a 20 week period in cynomolgus monkeys (2 males and 2 females per group) immunized 3 times intramuscularly. Serum IgE levels were determined in cynomolgus monkey IgE quantitative ELISA. The cynomolgus monkey serum was diluted in a 1:20 ratio. Cynomolgus IgE production standard curves were used. By mixing A with 450 IgE concentration was calculated by interpolation to a standard curve generated by nonlinear regression using a 4-parameter logistic curve fit. Results are mean ± standard deviation. Paired t assays with double tail hypotheses were used to identify statistical differences versus week 0: * P < 0.05, P < 0.01 and P < 0.001.
Detailed Description
The present disclosure relates to peptide immunogen constructs targeting the Extracellular Membrane Proximal Domain (EMPD) of membrane-bound IgE (or IgE EMPD). The disclosure also relates to compositions comprising such peptide immunogen constructs, methods of making and using such peptide immunogen constructs, and antibodies produced by hosts immunized with such peptide immunogen constructs.
The disclosed peptide immunogen constructs comprise about 20 or more amino acids. The peptide immunogen construct comprises a B cell epitope of 67 amino acid sequences from full length IgE EMPD (SEQ ID NO: 1). This B cell epitope may be linked through an optional heterologous spacer to a heterologous T helper cell (Th) epitope derived from a pathogen protein. The disclosed peptide immunogen constructs may stimulate the production of highly specific antibodies to IgE EMPD and may bind to recombinant IgE EMPD-containing protein γ1-em67, and/or IgE EMPD on B cells with mIgE. The disclosed peptide immunogen constructs are useful as a cost-effective, general-purpose immunotherapy for global patients with IgE-mediated allergic diseases, independent of the allergen.
The B cell epitope portion of the peptide immunogen construct has an amino acid sequence derived from the full-length IgE EMPD sequence (SEQ ID NO: 1). In some embodiments, the B cell epitope has a sequence comprising an internal intramolecular loop formed by endogenous cysteines (C18-C39) according to the numbering of the full length IgE EMPD sequence (SEQ ID NO: 1). In certain embodiments, the B cell epitope has the amino acid sequence of IgE EMPD-1-39 (SEQ ID NO: 5), igE EMPD-7-40 (SEQ ID NO: 6), igE EMPD-19-38 (SEQ ID NO: 8), or IgE EMPD-1-40 (SEQ ID NO: 9).
The peptide immunogen constructs of the present disclosure may contain heterologous Th epitope amino acid sequences derived from pathogen proteins (e.g., SEQ ID NOS: 59 to 87). In certain embodiments, the heterologous Th epitope is derived from a pathogen in nature, for example: diphtheria toxin (SEQ ID NO: 63), plasmodium falciparum (SEQ ID NO: 64), cholera toxin (SEQ ID NO: 66). In other embodiments, the heterologous Th epitope is an idealized artificial Th epitope derived from measles virus fusion proteins (MVF 1 to 5) or hepatitis B surface antigen (HBsAg 1 to 3) in the form of a single sequence (e.g., SEQ ID NOS: 60, 67, 72 and 73) or a combined sequence (e.g., SEQ ID NOS: 70, 69 and 71).
In some embodiments, the peptide immunogen construct comprises a B cell epitope from IgE EMPD linked to a heterologous T helper cell (Th) epitope through an optional heterologous spacer. An optional heterologous spacer is a molecule or chemical structure capable of linking two amino acids and/or peptides together. In certain embodiments, the spacer is a naturally occurring amino acid, a non-naturally occurring amino acid, or a combination thereof.
In certain embodiments, the peptide immunogen construct comprises a B cell antigenic site from IgE EMPD-1-40 (SEQ ID NO: 9) having more than about 20 amino acids linked by an optional heterologous spacer to a heterologous Th epitope derived from a pathogen protein (e.g., SEQ ID NO:59 to 87). In a specific embodiment, the peptide immunogen construct has the sequence of SEQ ID NO:88-95, 98-124 and 130.
The disclosure also relates to compositions comprising IgE EMPD peptide immunogen constructs. In some embodiments, the disclosed compositions comprise more than one IgE EMPD peptide immunogen construct. In certain embodiments, the compositions comprise a mixture of peptide immunogen constructs comprising B cell epitope portions of IgEEMPD-1-39 linked to different Th epitopes (e.g., any combination of SE Q ID NOs: 98-124) to encompass a broad genetic background for the patient. Compositions comprising a mixture of peptide immunogen constructs may result in a higher percentage of response after vaccination compared to compositions comprising only a single peptide immunogen construct to treat IgE-mediated allergic diseases.
The present disclosure also relates to pharmaceutical compositions, including vaccine dosage forms, for the treatment and/or prevention of IgE-mediated allergic diseases. In some embodiments, the pharmaceutical composition comprises the disclosed peptide immunogen constructs in the form of a stabilized immunostimulatory complex formed by mixing a CpG oligomer and a composition comprising the peptide immunogen complex to form by electrostatic binding. Such stabilized immunostimulatory complexes may further enhance the immunogenicity of the peptide immunogen construct. In some embodiments, the pharmaceutical composition comprises an adjuvant, such as a mineral salt, including alum gel (ALHYDROGEL), aluminum phosphate (ADJUPHOS), or a water-in-oil emulsion including MONTANIDE ISA 51 or 720.
The disclosure also relates to antibodies directed against the disclosed IgE EMPD peptide immunogen constructs. In particular, the peptide immunogen constructs of the present disclosure may stimulate the production of highly specific antibodies that cross-react with the IgE EMPD B cell epitope portion of the peptide immunogen construct. The disclosed antibodies bind to IgE EMPD with high specificity, but not very much (if any) to heterologous Th epitopes for immunogenicity enhancement, in sharp contrast to antibodies made using conventional proteins or other biological carriers for peptide immunogenicity enhancement. Thus, the disclosed peptide immunogen constructs can break the immune tolerance against self-antigens with high reactivity compared to other peptide or protein immunogens.
In certain embodiments, when the peptide immunogen construct is administered to a subject, the antibody is directed against and specifically binds to the IgE EMPD-1-52 amino acid sequence (SEQ ID NO: 2), igE EMPD-1-67 amino acid sequence (SEQ ID NO: 1), and fragments thereof (e.g., SEQ ID NOs:5 and 6). Highly specific antibodies made using the peptide immunogen constructs can cross-react with soluble IgE EMPD-containing peptides and proteins, igE EMPD-containing fusion peptides and proteins, γ1-em67, and/or IgE EMPD on B cells with membrane-bound IgE. The antibodies produced are capable of binding and cross-linking to IgE B Cell Receptors (BCR) on B lymphocytes expressing mIgE. Such cross-linking can induce cytolytic effects, such as apoptosis and Antibody Dependent Cellular Cytotoxicity (ADCC), which result in reduced serum IgE production.
Based on their unique features and properties, the disclosed antibodies can provide a general immunotherapeutic approach to treat IgE-mediated allergic diseases, regardless of the pathogenic allergen species.
The disclosure also relates to methods of making the disclosed peptide immunogen constructs, compositions and antibodies. The disclosed methods provide for low cost manufacture and quality control of peptide immunogen constructs and compositions containing such constructs, which are useful in methods of treating IgE-mediated allergic diseases, regardless of the pathogenic allergen species.
The present disclosure also includes methods for treating and/or preventing IgE-mediated allergic diseases, regardless of the pathogenic allergen species, using the disclosed peptide immunogen constructs and/or antibodies directed against such peptide immunogen constructs. In some embodiments, a method for treating and/or preventing IgE-mediated allergic diseases comprises administering to a host a composition comprising the disclosed peptide immunogen constructs. In certain embodiments, the compositions used in the methods comprise a disclosed peptide immunogen construct in the form of a stabilized immunostimulatory complex formed by electrostatic binding using negatively charged oligonucleotides (e.g., cpG oligomers), which may be further supplemented with an adjuvant for administration to IgE-mediated allergic disease patients. The disclosed methods also include administration of the peptide immunogen constructs to a dosage regimen, dosage form and route of administration to a host at risk of IgE-mediated allergic disease or that has developed.
The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described. All references or portions of references cited in this application are expressly incorporated herein by reference in their entirety for any purpose.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this application belongs. The singular terms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. Similarly, the word "or" is intended to include "and" unless the context clearly indicates otherwise. Thus, "comprising A or B" is meant to include A, or B, or A and B. It should further be understood that all amino acid sizes and all molecular weights or molecular mass values for a given polypeptide are approximate and are provided for descriptive purposes. However, methods and materials similar or equivalent to those described herein can be used in the practice or testing of the disclosed methods, suitable methods and materials described below. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including an explanation of the terms, will control. In addition, the materials, methods, and embodiments disclosed herein are illustrative only and are not intended to be limiting.
IgE EMPD peptide immunogen constructs
The present disclosure provides peptide immunogen constructs comprising B cell epitopes having an amino acid sequence from IgE EMPD, the B cell epitopes being covalently linked to heterologous T helper cell (Th) epitopes either directly or through optional heterologous spacers.
The term "IgE EMPD peptide immunogen construct" or "peptide immunogen construct" as used herein refers to a B cell epitope having about 20 or more amino acid residues comprising (a) 67 amino acid sequences from full-length IgE EMPD (SEQ ID NO: 1); (b) a heterologous Th epitope; and (c) optionally a peptide of a heterologous spacer.
In certain embodiments, the IgE EMPD peptide immunogen construct may be represented by the formula:
(Th) m -(A) n - (IgE EMPD fragment) -X
Or (b)
(IgE EMPD fragment) - (A) n -(Th) m -X
Or (b)
(Th) m -(A) n - (IgE EMPD fragment) - (a) n -(Th) m -X
Wherein the method comprises the steps of
Th is a heterologous T helper epitope;
a is a heterologous spacer;
(IgE EMPD fragment) is a B cell epitope from IgE EMPD having about 20 to about 40 amino acid residues;
x is alpha-COOH or alpha-CONH of amino acid 2
m is 1 to about 4; and
n is 0 to about 10.
The IgE EMPD peptide immunogen constructs of the present disclosure were designed and selected based on a number of basic principles. Several of the rationales include the use of IgE EMPD peptide immunogen constructs:
i. Is itself non-immunogenic in that it is a self-molecule;
immunogenicity can be achieved by protein carriers or potent T helper epitopes;
when rendered immunogenic and administered to a host:
a. eliciting high titer antibodies against IgE EMPD peptide sequences (B cell epitopes) but not against protein carriers or T helper cell epitopes;
b. disruption of immune tolerance in immunized hosts and production of highly specific antibodies cross-reactive with IgE EMPD (SEQ ID NO: 1) expressed by mI gE.Fc L Recombinant proteins purified from CHO cells of (A) or by encoding mIgE. Fc L B cell (e.g. Ramos) cell membrane transfected with mIgE;
c. in vitro production of highly specific antibodies capable of inducing Antibody Dependent Cellular Cytotoxicity (ADCC) and apoptosis of IgE-expressing B lymphocytes (example 6); and
d. after primary and booster immunization with allergen challenge, highly specific antibodies can be produced, which can lead to in vivo reduction of basal levels of IgE in blood, as well as significant reduction of antigen-specific IgE levels (examples 8 to 11).
The disclosed IgE EMPD peptide immunogen constructs and dosage forms thereof are effective as vaccines to reduce or eliminate IgE-mediated allergic pathology in IgE-mediated allergic disease patients.
The various components of the disclosed IgE EMPD peptide immunogen constructs are described in further detail below.
B cell epitopes of IgE EMPD
The present disclosure relates to novel peptide compositions for the production of high titer polyclonal antibodies specific for IgE EMPD peptides that cross-react with membrane-bound IgE expressed on human B cells that differentiate committed to IgE secretion. With reasonable design efforts, the site-specificity of the peptide composition minimizes the production of antibodies directed against unrelated sites on the carrier protein.
The term "IgE" as used herein refers to any form of immunoglobulin E, including secreted IgE, membrane-bound IgE, and fragments thereof. Figure 2A illustrates IgE secretion and membrane-bound forms.
The term "mIgE" is used herein to refer in particular to membrane bound forms of IgE and fragments thereof. In some embodiments, the mIgE is a membrane bound form of IgE in humans having an amino acid sequence like Ig epsilon chain form 2 (fragment) of chain C region as reported under the number PH 1215. Figure 2A illustrates membrane bound IgE (right).
The term "IgE EMPD" as used herein refers to the membrane-associated IgE (mIgE) cell outer membrane proximal domain (EMPD) and fragments thereof. IgE EMPD is also known as C epsilon mX, which is located between the CH4 domain and the carboxy-terminal membrane-anchored transmembrane peptide, and is present only on mIgE B cells. The EMPD of IgE results from the alternative splicing of epsilon RNA transcripts 156bp upstream of the splice acceptor site used for the "short" isoform. The full length "long" EMPD isoform of human IgE is 67 amino acids in length (SEQ ID NO: 1), which comprises 52 amino acids that are not present in the "short" isoform (SEQ ID NO: 2). Figure 2A illustrates membrane bound IgE with an enlarged EMPD moiety. The amino acid sequences of full-length IgE EMPD (SEQ ID NO: 1) and fragments thereof (SEQ ID NO:2-58 and 127) are shown in Table 1.
IgE EMPD comprises an intramolecular cyclic structure between endogenous cysteines (C18-C39) based on the amino acid numbering of 67 amino acids and 52 amino acid sequences (SEQ ID NOs: 1 and 2, respectively). The intramolecular cyclic structure of IgE is illustrated in fig. 9.
The B cell epitope of the IgE EMP D peptide immunogen construct comprises the intramolecular cyclic structure of IgE EMPD, or a portion thereof. In certain embodiments, the B cell epitope comprises about 20 to about 40 amino acids of IgE EMPD.
In some embodiments, the amino acid sequence of the B cell epitope portion of the IgE EMPD peptide immunogen construct comprises about 20 to about 40 amino acid residues from full length IgE EMPD (SEQ ID NO: 1). In certain embodiments, the B cell epitope comprises an amino acid sequence from the internal intramolecular loop structure of IgE EMPD formed from endogenous cysteines (C18-C39), according to the numbering of full length IgE EMPD (SEQ ID NO: 1). In a specific embodiment, the sequence of the B cell epitope ends at the carboxy terminus of the intramolecular loop structure of IgE EMPD with arginine (R) at residue 38, cysteine (C) at residue 39, or histidine (H) at residue 40.
In some embodiments, as shown in Table 1, the B cell epitope has the amino acid sequence of IgE EMPD-1-39 (SEQ ID NO: 5), igE EMPD-7-40 (SEQ ID NO: 6), igE EMPD-19-38 (SEQ ID NO: 8), or IgE EMPD-1-40 (SEQ ID NO: 9).
IgE EMPD fragments of the present disclosure also include IgE EMPD peptides (SEQ ID NOs: 5, 6, 8 and 9) and immunologically functional analogs or homologs thereof over 20 amino acid fragments. Functional immune analogs or homologs of IgE EMPD peptides and more than 20 amino acid fragments thereof include variants that retain substantially the same immunogenicity as the original peptide. The epidemic functional analog may have conservative substitutions at amino acid positions, total charge changes, covalent linkages to other functional groups, or addition, insertion or deletion of amino acids, and/or any combination thereof.
b. Heterologous T helper cell epitope (Th epitope)
The present disclosure provides peptide immunogen constructs comprising B cell epitopes from IgE EMPD covalently linked to heterologous T helper cell (Th) epitopes either directly or through optional heterologous spacers.
Heterologous Th epitopes in IgE EMPD peptide immunogen constructs enhance the immunogenicity of IgE EMPD fragments, which through rational design promotes the production of specific high-potency antibodies against optimized B-cell epitopes (i.e. IgE EMPD fragments).
The term "heterologous" as used herein refers to an amino acid sequence derived from an amino acid sequence that is not part of or homologous to the IgE EMPD wild-type sequence. Thus, a heterologous Th epitope is a Th epitope derived from an amino acid sequence that is not naturally present in IgE EMPD (i.e., the Th epitope is not self-derived to IgE EMPD). Because Th epitopes are heterologous to IgE EMPD, when heterologous Th epitopes are covalently linked to IgE EMPD fragments, the native amino acid sequence of IgE EMPD does not extend in the amino-or carboxy-terminal direction.
The heterologous Th epitope of the present disclosure may be any Th epitope that does not have the amino acid sequence naturally present in IgE EMPD. Th epitopes may also have promiscuous binding motifs to MHC class II molecules of various species. In certain embodiments, th epitopes comprise multiple promiscuous MHC class II binding motifs to allow maximum activation of T helper cells, resulting in initiation and modulation of immune responses. Preferred Th epitopes are themselves non-immunogenic (i.e. antibodies raised against Th epitopes with little, if any, igE EMPD peptide immunogen constructs) thus allowing for a very focused immune response against the target B cell epitopes of the IgE EMPD fragment.
Th epitopes of the present disclosure include, but are not limited to, amino acid sequences derived from foreign pathogens, as exemplified in Table 2 (SEQ ID NOS: 59-87). In addition, th epitopes include idealized artificial Th epitopes and combinations of idealized artificial Th epitopes (e.g., SEQ ID NOS: 60 and 67-73). The heterologous Th epitope peptides are presented in a combined sequence (e.g., SEQ ID NO: 68-71) comprising a mixture of amino acid residues represented at specific positions within the peptide backbone based on variable residues of homologs of the specific peptide. A collection of combinatorial peptides can be synthesized in a single process using a mixture of selected protected amino acids added at specific positions during the synthesis process, rather than one specific amino acid. Such a combination of heterologous Th epitope peptide sets may allow for broad coverage of Th epitopes for animals with different genetic backgrounds. Representative combinatorial sequences of heterologous Th epitope peptides comprise SEQ ID NOs: 68-71. The Th epitope peptides of the present invention provide broad reactivity and immunogenicity to animals and patients from genetically diverse populations.
IgE EMPD peptide immunogen constructs comprising Th epitopes may be produced simultaneously in a single solid phase peptide synthesis in tandem with IgE EMPD fragments. Th epitopes may also include immune analogues of Th epitopes. Immune Th analogs include immune enhancing analogs, cross-reactive analogs, and fragments of any of these Th epitopes, sufficient to enhance or stimulate an immune response to IgE EMPD fragments.
Functional immune analogs of Th epitope peptides are also effective and are included as part of the present invention. Functional immune Th analogs may comprise conservative substitutions, additions, deletions and insertions of from 1 to about 5 amino acid residues in a Th epitope that do not substantially alter the Th stimulating function of the Th epitope. Conservative substitutions, additions and insertions may be made using natural or unnatural amino acids as described above for IgE EMPD fragments. Table 2 identifies another variant of a functional analogue of a Th epitope peptide. Specifically, SEQ ID NOs of MvF1 and MvF Th: 60 and 67 are SEQ ID NOs of MvF4 and MvF 5: 70 and 72, because the amino acid backbones are distinguished by deletion (SEQ ID NOS: 60 and 67) or insertion (SEQ ID NOS: 70 and 72) of two amino acids each at the amino-and carboxy-terminus. The difference between these two series of similar sequences does not affect the function of Th epitopes contained in these sequences. Thus, functional immune Th analogs include versions of Th epitopes derived from measles virus fusion proteins MvF1-4 Ths (SEQ ID NOS: 60, 67, 68, 70 and 72) and from hepatitis surface protein HBsAg 1-3 Ths (SEQ ID NOS: 69, 71 and 73).
The Th epitope in the IgE EMPD peptide immunogen construct may be covalently linked to the amino-or carboxy-terminus of the IgE EMPD peptide fragment. In some embodiments, the Th epitope is covalently linked to the amino terminus of the IgE EMPD peptide fragment. In other embodiments, the Th epitope is covalently linked to the carboxy terminus of the IgE EMPD peptide fragment. In certain embodiments, more than one Th epitope is covalently linked to an IgE EMPD fragment. When more than one Th epitope is linked to an IgE EMPD fragment, each Th epitope may have the same amino acid sequence or different amino acid sequences. In addition, when more than one Th epitope is linked to an IgE EMPD fragment, the Th epitopes may be arranged in any order. For example, a Th epitope may be linked serially to the amino terminus of an IgE EMPD fragment, or to the carboxy terminus of an IgE EMPD fragment, or when different Th epitopes are covalently linked to the carboxy terminus of an IgE EMPD fragment, the Th epitope may be covalently linked to the amino terminus of an IgE EMPD fragment. The arrangement of Th epitopes relative to IgE EMPD fragments is not limited.
In some embodiments, the Th epitope is directly covalently linked to the IgE EMPD fragment. In other embodiments, the Th epitope is covalently linked to the IgE EMPD fragment through a heterologous spacer as described in further detail below.
c. Heterologous spacers
The disclosed IgE EMPD peptide immunogen constructs optionally comprise a heterologous spacer that covalently links B-cell epitopes from IgE EMPD to heterologous T-helper (Th) epitopes.
As mentioned above, the term "heterologous" refers to an amino acid sequence derived from an amino acid sequence that is not part of or homologous to the native form of IgE EMPD sequence. Thus, when a heterologous spacer is covalently linked to a B cell epitope from IgE EMPD, the native amino acid sequence of IgE EMPD does not extend in the amino-or carboxy-terminal direction, as the spacer is heterologous to the IgE EMPD sequence.
A spacer is a molecule or chemical structure that is capable of linking two amino acids and/or peptides together. The length or polarity of the spacers may vary depending on the application. The spacer linkages may be linked by amide or carboxyl groups, but other functional groups are also possible. The spacer may include chemical compounds, naturally occurring amino acids, or non-naturally occurring amino acids.
The spacer may provide structural features to the IgE EMPD peptide immunogen construct. Structurally, the spacer provides physical separation of Th epitopes from B cell epitopes of IgE EMPD fragments. Physical separation by the spacer can disrupt any artificial secondary structure created by linking the Th epitope to the B cell epitope. In addition, interference between Th cell and/or B cell responses can be eliminated by physical separation of epitopes of the spacer. In addition, the spacer may be designed to create or modify the secondary structure of the peptide immunogen construct. For example, spacers can be designed to act as flexible hinges to enhance the separation of Th and B cell epitopes. The flexible hinge spacer may also allow for more efficient interaction between the presented peptide immunogen and appropriate Th and B cells to enhance immune responses to Th and B cell epitopes. An illustration of the sequence encoding the flexible hinge is found in the immunoglobulin heavy chain hinge region, which is typically proline rich. The use of the sequence Pro-Pro-Xaa-Pro-Xaa-Pro (SEQ ID NO: 128) provides a particularly useful flexible hinge as a spacer, where Xaa is any amino acid, preferably aspartic acid.
The spacer may also provide functional features to the IgE EMPD peptide immunogen construct. For example, the spacer may be designed to alter the overall charge of the IgE EMPD peptide immunogen construct, which may affect the solubility of the peptide immunogen construct. Furthermore, altering the overall charge of an IgE EMPD peptide immunogen construct may affect the ability of the peptide immunogen construct to bind to other compounds and agents. As discussed in further detail below, igE EMPD peptide immunogen constructs may form stable immunostimulatory complexes with highly charged oligonucleotides (e.g., cpG oligomers) by electrostatic binding. The total charge of the IgE EMPD peptide immunogen construct is important for the formation of these stable immunostimulatory complexes.
Chemical compounds that may be used as spacers include, but are not limited to, (2-aminoethoxy) acetic acid (AEA), 5-aminopentanoic acid (AVA), 6-aminocaproic acid (Ahx), 8-amino-3, 6-dioxaoctanoic acid (AEEA, mini-PEG 1), 12-amino-4, 7, 10-trioxadodecanoic acid (mini-PEG 2), 15-amino-4, 7, 10, 13-tetraoxapentadecanoic acid (mini-PEG 3), trioxatadecan-succinic acid (Ttds), 12-aminododecanoic acid, fmoc-5-amino-3-oxopentanoic acid (O1 Pen), and the like.
Naturally occurring amino acids include alanine, arginine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, and valine.
Non-naturally occurring amino acids include, but are not limited to, epsilon-N lysine, beta-alanine, ornithine, norleucine, norvaline, hydroxyproline, thyroxine, gamma-aminobutyric acid, homoserine, citrulline, aminobenzoic acid, 6-aminocaproic acid (Aca; 6-aminocaproic acid), 3-mercaptopropionic acid (MPA), 3-nitrotyrosine, pyroglutamic acid, and the like.
The spacer in the IgE EMPD peptide immunogen construct may be covalently linked to the Th epitope and the amino-or carboxy-terminus of the IgE EMPD peptide. In some embodiments, the spacer is covalently linked to the carboxy terminus of the Th epitope and the amino terminus of the IgE EMPD peptide. In other embodiments, the spacer is covalently linked to the carboxy terminus of the IgE EMPD peptide and the amino terminus of the Th epitope. In certain embodiments, more than one spacer may be used, for example, when more than one Th epitope is present in the peptide immunogen construct. When more than one spacer is used, each spacer may be the same or different from each other. In addition, when more than one Th epitope is present in the peptide immunogen construct, the Th epitopes may be separated by a spacer, which may be the same or different, and the Th epitopes are separated from the B cell epitopes by a spacer. The arrangement of the spacer with respect to the Th epitope or IgE EMPD fragment is not limited.
In certain embodiments, the heterologous spacer is a naturally occurring amino acid or a non-naturally occurring amino acid. In other embodiments, the spacer comprises more than one naturally occurring or non-naturally occurring amino acid. In particular embodiments, the spacer is Lys-, gly-, lys-Lys-Lys-, (α, ε -N) Lys or ε -N-Lys-Lys-Lys (SEQ ID NO: 129).
Specific embodiments of IgE EMPD peptide immunogen constructs
In certain embodiments, igE EMPD peptide immunogen constructs may be represented by the following formula:
(Th) m -(A) n - (IgE EMPD fragment) -X
Or (b)
(IgE EMPD fragment))-(A) n -(Th) m -X
Or (b)
(Th) m -(A) n - (IgE EMPD fragment) - (a) n -(Th) m -X
Wherein the method comprises the steps of
Th is a heterologous T helper epitope;
a is a heterologous spacer;
(IgE EMPD fragment) is a B cell epitope from IgE EMPD having about 20 to about 40 amino acid residues;
x is alpha-COOH or alpha-CONH of amino acid 2
m is 1 to about 4; and
n is 0 to about 10.
In certain embodiments, the heterologous Th epitope in the IgE EMPD peptide immunogen construct has a sequence selected from the group consisting of SEQ ID NOs: 59-87, and combinations thereof, as shown in table 2. In some embodiments, the IgE EMPD peptide immunogen construct comprises more than one Th epitope.
In certain embodiments, the optional heterologous spacer is any one selected from the group consisting of Lys-, gly-, lys-Lys-Lys-, (α, ε -N) Lys, ε -N-Lys-Lys-Lys (SEQ ID NO: 129), and combinations thereof. In a specific embodiment, the heterologous spacer is ε -N-Lys-Lys-Lys-Lys (SEQ ID NO: 129).
In certain embodiments, the IgE EMPD fragment has a sequence from SEQ ID NO:1 or 2 from about 20 to about 40 amino acid residues of IgE EMPD. In a specific embodiment, the IgE EMPD fragment comprises an amino acid sequence from the internal intramolecular ring structure of IgE EMPD formed from endogenous cysteines (C18-C39) according to the numbering of full length IgE EMPD (SEQ ID NO: 1). In specific embodiments, the IgE EMPD fragment has the amino acid sequence of IgE EMPD-1-39 (SEQ ID NO: 5), igE EMPD-7-40 (SEQ ID NO: 6), igE EMPD-19-38 (SEQ ID NO: 8) or IgE EMPD-1-40 (SEQ ID NO: 9) as shown in Table 1.
In certain embodiments, the IgE EMPD peptide immunogen construct has a sequence selected from the group consisting of SEQ ID NOs: 88-130, as shown in table 3. In specific embodiments, the IgE EMP D peptide immunogen construct has a sequence selected from the group consisting of SEQ ID NOs: 88-95, 98-124 and 130.
e. Variants, homologs and functional analogs
Variants and analogs of the above immunogenic peptides may also be used which induce and/or cross-react with antibodies, which are preferred epitopes for IgE EMPD. Analogs (including alleles, species and induced variants) typically differ from naturally occurring peptides in one, two or several positions, typically due to conservative substitutions. Analogs typically exhibit at least 80 or 90% sequence identity to the native peptide. Some analogs also include modifications of unnatural amino acids or amino-or carboxy-terminal amino acids at one, two, or several positions.
Variants that are functional analogs can have conservative substitutions at amino acid positions, total charge changes, covalent linkages to other functional groups, or addition, insertion, or deletion of amino acids, and/or any combination thereof.
Conservative substitutions refer to the substitution of one amino acid residue for another amino acid residue having similar chemical properties. For example, nonpolar (hydrophobic) amino acids include alanine, leucine, isoleucine, valine, proline, phenylalanine, tryptophan, and methionine; polar neutral amino acids include glycine, serine, threonine, cysteine, tyrosine, asparagine, and glutamine; positively charged (basic) amino acids include arginine, lysine and histidine; and negatively charged (acidic) amino acids include aspartic acid and glutamic acid.
In a particular embodiment, the functional analog has at least 50% identity to the original amino acid sequence. In another embodiment, the functional analog has at least 80% identity to the original amino acid sequence. In yet another embodiment, the functional analog has at least 85% identity to the original amino acid sequence. In yet another embodiment, the functional analog has at least 90% identity to the original amino acid sequence.
Variants also include changes in phosphorylated residues. For example, variants may include different residues within the peptide that is phosphorylated. Variant immunogenic IgE EMPD peptides may also include pseudophosphorylated peptides. Pseudo-phosphorylated peptides are produced by substitution of one or more phosphorylated serine, threonine and tyrosine residues of IgE EMPD peptides with acidic amino acid residues (e.g., glutamic acid and aspartic acid).
Composition and method for producing the same
The present disclosure also provides compositions comprising the disclosed IgE EMPD immunogen constructs.
a. Peptide composition
The compositions comprising the disclosed IgE EMPD peptide immunogen constructs may be in liquid or solid form. The liquid composition may include water, buffers, solvents, salts, and/or any other acceptable agent that does not alter the structural or functional properties of the IgE EMPD peptide immunogen construct. The peptide composition may contain one or more of the disclosed IgE EMPD peptide immunogen constructs.
b. Pharmaceutical composition
The disclosure also relates to pharmaceutical compositions comprising the disclosed IgE EMPD peptide immunogen constructs.
The pharmaceutical composition may contain carriers and/or other additives in a pharmaceutically acceptable delivery system. Thus, the pharmaceutical composition may contain a pharmaceutically effective dose of the IgE EMPD peptide immunogen construct in combination with pharmaceutically acceptable carriers, adjuvants and/or other excipients (e.g., diluents, additives, stabilizers, preservatives, co-solvents, buffers, etc.).
The pharmaceutical composition may contain one or more adjuvants whose function is to accelerate, prolong or enhance the immune response against the IgE EMPD peptide immunogen construct without having any specific antigenic effect itself. Adjuvants used in pharmaceutical compositions may include oils, oil emulsions, aluminum salts, calcium salts, immunostimulatory complexes, bacterial and viral derivatives, virosomes, carbohydrates, cytokines, polymer microparticles. In certain embodiments, the adjuvant may be selected from alum (potassium aluminum phosphate), aluminum phosphate (e.g., ADJU-) Aluminum hydroxide (e.g.)>) Calcium phosphate, freund's incomplete adjuvant (IFA), freund's complete adjuvant, MF59, adjuvants 65, lipovant, ISCOM, liposyn, saponins, squalene, L121, and- >Monophosphoryl lipid a (MPL), quil a, QS21, ++>ISA 35, ISA 50V, ISA V2, ISA 51, ISA 206, ISA 720, liposomes, phospholipids, peptidoglycans, lipopolysaccharides (LPS), ASO1, ASO2, ASO3, ASO4, AF03, lipophilic phospholipids (lipid a), gamma inulin, algal inulin (algammulin), dextran, glucomannan, galactomannan, fructopolysaccharides, xylans, dimethyl dioctadecyl ammonium bromide (DDA), and other adjuvants and emulsifiers.
In some embodiments, the pharmaceutical composition contains Montanide TM ISA 51 (oleaginous adjuvant composition consisting of vegetable oil and manna-diolide oleate for the manufacture of water-in-oil emulsions),80 (also known as polysorbate 80 or polyoxyethylene (20) sorbitan monooleate), cpG oligonucleotides, and/or any combination thereof. In other embodiments, the pharmaceutical composition is a water-in-oil-in-water (i.e., w/o/w) emulsion with Emulsig or Emulsig D as an adjuvant.
The pharmaceutical composition may also include pharmaceutically acceptable additives or excipients. For example, the pharmaceutical composition may contain antioxidants, binders, buffers, bulking agents, carriers, chelating agents, colorants, diluents, disintegrants, emulsifiers, fillers, gelling agents, pH buffers, preservatives, co-solvents, stabilizers, and the like.
The pharmaceutical composition may be formulated into a sustained or sustained release dosage form. In addition, the pharmaceutical compositions may be formulated for induction of systemic or local mucosal immunity by encapsulation of the immunogen and co-administration with the microparticles. Such delivery systems are readily determinable by one of ordinary skill in the art.
The pharmaceutical compositions may be formulated as injectables, either as liquid solutions or suspensions. Liquid carriers containing IgE EMPD peptide immunogen constructs may also be prepared prior to injection. The pharmaceutical composition may be administered using any suitable method of use, e.g., i.d., i.v., i.p., i.m., intranasal, oral, subcutaneous, etc., and may be administered in any suitable delivery device. In certain embodiments, the pharmaceutical composition may be formulated for intravenous, subcutaneous, intradermal, or intramuscular administration. Pharmaceutical compositions suitable for other modes of administration may also be prepared, including oral and intranasal applications.
The pharmaceutical compositions may also be formulated in suitable dosage unit form. In some embodiments, the pharmaceutical composition contains about 0.1 μg to about 1mg IgE EMPD peptide immunogen construct per kilogram body weight. The effective dosage of the pharmaceutical composition depends on many different factors including the mode of administration, the target, the physiological state of the patient, whether the patient is a human or animal, other drugs administered, and whether the treatment is for prophylaxis or treatment. Typically, the patient is a human, but non-human mammals including transgenic mammals can also be treated. When delivered in multiple doses, the pharmaceutical composition can be conveniently divided into appropriate amounts in each dosage unit form. The dose administered will depend on the age, weight and general health of the subject as is well known in the therapeutic arts.
In some embodiments, the pharmaceutical composition contains more than one IgE EMPD peptide immunogen construct. Pharmaceutical compositions containing mixtures of more than one IgE EMPD peptide immunogen constructs allow synergistic enhancement of the immunopotency of the construct. Pharmaceutical compositions containing more than one IgE EMPD peptide immunogen construct may be more effective in larger genetic populations due to extensive MHC class II coverage, thus providing improved immune responses against IgE EMPD peptide immunogen constructs.
In some embodiments, the pharmaceutical composition contains a polypeptide selected from the group consisting of SE Q ID NO:88-95, 98-124 and 130 (Table 3), and homologs, analogs and/or combinations thereof.
In certain embodiments, igE EMPD peptide immunogen constructs (SEQ ID NOS: 107 and 108) having heterologous Th epitopes derived from MVF and HBsAg in combination (SEQ ID NOS: 68 and 69) are mixed in equimolar ratios for use in vaccine dosage forms to allow maximum coverage of vaccine-receiving host populations having different genetic backgrounds. Synergistic enhancement was observed in the peptide compositions of the present invention in IgE EMPD G1-C39 and A7-C40 immunogen constructs (SEQ ID NOS: 88 to 95), and the antibody response elicited by such constructs (e.g., SEQ ID NOS: 95) was largely (> 90%) focused on the desired cross-reactivity against the B cell epitope peptide of IgE EMPD (SEQ ID NO: 2) with little, if any, against the heterologous Th epitope for immunogenicity enhancement (example 7, table 7). This is in sharp contrast to antibodies made with conventional proteins (e.g., KLH) or other bioprotein vectors used for peptide antigenicity enhancement.
In other embodiments, a pharmaceutical composition comprising a peptide composition, such as a mixture of IgE EMPD peptide immunogen constructs contacted with a mineral salt as an adjuvant, including alum gel (alum) or aluminum phosphate (ajuphos), forms a suspension vaccine dosage form for administration to a vaccine host.
Pharmaceutical compositions containing IgE EMPD peptide immunogen constructs may be used to elicit an immune response and produce antibodies in a host following administration.
c. Immunostimulatory complexes
The present disclosure also relates to pharmaceutical compositions containing IgE EMPD peptide immunogen constructs that form immunostimulatory complexes with CpG oligonucleotides. Such immunostimulatory complexes are particularly suitable as adjuvants and peptide immunogen stabilizers. The immunostimulatory complex is in particulate form, which is effective to present IgE EMPD peptide immunogens to cells of the immune system to generate an immune response. The immunostimulatory complexes may be formulated as suspensions for parenteral administration. The immunostimulatory complexes may also be formulated in the form of a w/o emulsion as a suspension in combination with a mineral salt or in situ gel polymer for efficient delivery of IgE EMPD peptide immunogens to cells of the host immune system following parenteral administration.
The stabilized immunostimulatory complexes may be formed by complexing the igempd peptide immunogen construct with an anionic molecule, oligonucleotide, polynucleotide, or combination thereof by electrostatic binding. The stabilized immunostimulatory complexes may be incorporated into a pharmaceutical composition as an immunogen delivery system.
In certain embodiments, igE EMPD peptide immunogen constructs are designed to comprise a cationic moiety that has a positive charge at a pH ranging from 5.0 to 8.0. The net charge calculation of the cationic portion of the IgE EMPD peptide immunogen constructs or mixtures of constructs is based on the fact that each lysine (K), arginine (R) or histidine (H) has a +1 charge, each aspartic acid (D) or glutamic acid (E) has a-1 charge, and the charge of the other amino acids in the sequence is 0. The charge of the cationic moiety in the IgE EMPD peptide immunogen construct is added and expressed as the net average charge. Suitable peptide immunogens have a cationic moiety with a net average positive charge of +1. Preferably, the peptide immunogen has a net positive charge in the range of greater than +2. In some embodiments, the cationic portion of the IgE EMPD peptide immunogen construct is a heterologous spacer. In certain embodiments, when the spacer sequence is (. Alpha.,. Epsilon. -N) Lys, epsilon. -N-Lys-Lys-Lys-Lys (SEQ ID NO: 129), the cationic portion of the IgE EMPD peptide immunogen construct has a charge of +4.
As used herein, an "anionic molecule" refers to any molecule that has a negative charge at a pH in the range of 5.0 to 8.0. In certain embodiments, the anionic molecule is an oligomer or polymer. The net negative charge on the oligomer or polymer is calculated based on the charge of-1 per phosphodiester or phosphorothioate group in the oligomer. Suitable anionic oligonucleotides are single-stranded DNA molecules having 8 to 64 nucleotide bases, the number of repetitions of the CpG motif being in the range of 1 to 10. Preferably, the CpG immunostimulatory single stranded DNA molecule contains 18 to 48 nucleotide bases and the number of CpG motifs repeats is in the range of 3 to 8.
More preferably, the anionic oligonucleotide may be of formula 5' X 1 CGX 2 3' wherein C and G are unmethylatedThe method comprises the steps of carrying out a first treatment on the surface of the And X is 1 Is selected from the group consisting of A (adenine), G (guanine) and T (thymine); and X is 2 Is C (cytosine) or T (thymine). Alternatively, the anionic oligonucleotide may be of formula 5' (X) 3 ) 2 CG(X 4 ) 2 3' wherein C and G are unmethylated; and X is 3 Is selected from the group consisting of A, T or G; and X is 4 Is C or T.
The resulting immunostimulatory complex is in particulate form, typically in the range of 1-50 microns in size, and is a function of many factors, including the relative charge stoichiometry and molecular weight of the interacting components. The microparticle immunostimulatory complexes have the advantage of providing adjuvanting and up-regulation of specific immune responses in vivo. In addition, the stabilized immunostimulatory complexes are suitable for use in preparing pharmaceutical compositions by a variety of methods including water-in-oil emulsions, mineral salt suspensions, and polymeric gels.
The present disclosure also relates to pharmaceutical compositions, including vaccine dosage forms, for the treatment and prevention of IgE-mediated allergic diseases. In some embodiments, the pharmaceutical composition comprises a stabilized immunostimulatory complex formed by mixing a CpG oligomer and a peptide composition comprising a mixture of IgE EMPD peptide immunogen constructs (e.g., SEQ ID NOs: 88-95, 98-124 and 130) to further enhance the immunogenicity of the IgE EMPD peptide by electrostatic binding and elicit a peptide sequence directed against SEQ ID NO:1 or 2 to bind to B cells expressing mIgE and induce Antibody Dependent Cellular Cytotoxicity (ADCC) and apoptosis thereof (example 6).
In yet another embodiment, the pharmaceutical composition contains a mixture of IgE EMPD peptide immunogen constructs (e.g., any combination of SEQ ID NOs: 8-90, 94, 95, 98-124 and 130) that form a stabilized immunostimulatory complex with the CpG oligomer, the immunostimulatory complex being admixed with a mineral salt (including alum gel (ALHYDROGEL) or aluminum phosphate (ADJUPHOS)) as an adjuvant, optionally having a high safety factor, to form a suspension vaccine dosage form for administration to a host receiving the vaccine.
Antibodies to
The disclosure also provides antibodies raised using IgE EMPD peptide immunogen constructs.
The present disclosure provides IgE EMPD peptide immunogen constructs and dosage forms thereof that are cost effective in manufacture, optimally designed to elicit high titers of antibodies targeting membrane-bound IgE that can utilize high reactivity in immunized hosts to disrupt immune tolerance against autoantigens. Antibodies produced using IgE EMPD peptide immunogen constructs have high affinity for IgE-EMPD proteins, whether soluble peptides, fusion proteins, or IgE presented on IgE-bearing B cells. The antibodies produced are capable of binding and cross-linking IgE BCR on IgE-expressing B lymphocytes to induce cytolytic effects, such as apoptosis and ADCC. Apoptosis depletion of B lymphocytes of membrane-bound IgE can further result in reduced serum IgE production. Thus, targeting human migb cells with IgE EMPD-specific antibodies that induce apoptosis produced using the disclosed IgE EMPD peptide immunogen constructs and formulations thereof can provide novel therapies and vaccines against IgE-mediated allergic diseases.
In some embodiments, the IgE EMPD peptide immunogenic constructs used to elicit antibodies comprise a mixture of IgE EMPD peptides having B cell epitopes containing 20 to 40 amino acids (e.g., igE EMPD G1-C39 (SEQ ID NO: 5), igE EMPD A7-H40 (SEQ ID NO: 6), igE EMPD H19-R38 (SEQ ID NO: 8) and IgE EMPD 1-H40 (SEQ ID NO: 9) that encompass a ring structure within the central molecule derived from the IgE EMPD peptide (SEQ ID NO: 1) linked by an optional spacer to a heterologous Th epitope derived from a pathogen protein, such as Measles Virus Fusion (MVF) protein (SEQ ID NO: 73) or other (SEQ ID NO:59 to 87). B cell epitopes and Th epitopes of IgE EMPD peptide immunogen constructs act together to stimulate the production of highly specific antibodies that cross-react with IgE EMPD1-52 peptide (SEQ ID NO: 2), igE EMPD 1-67 protein (SEQ ID NO: 1), whether recombinant IgE EMPD-containing proteins (e.g., purified from a stable Flp-In CHO cell line transfected with recombinant DNA encoding the Fc portion of human IgG1 and IgE EMPD (. Gamma.1-em 67) of human membrane-bound IgE) or on the cell membrane of cells bearing membrane-bound IgE (e.g., purified using a recombinant DNA encoding mIgE. Fc) L A recombinant DNA transfected Ramos cell line).
Conventional methods to enhance peptide immunogenicity, such as by chemically coupling carrier proteins, such as Keyhole Limpet Hemocyanin (KLH) or other carrier proteins, such as Diphtheria Toxoid (DT) and Tetanus Toxoid (TT) proteins, typically result in the production of large amounts of antibodies directed against the carrier proteins. Thus, the main disadvantage of such peptide-carrier protein vaccines is that most (> 90%) of the antibodies produced using such immunogens are non-functional antibodies to the carrier proteins KLH, DT or TT which can lead to epitope suppression.
Unlike traditional methods to enhance peptide immunogenicity, antibodies generated using the disclosed IgE EMPD peptide immunogen constructs can bind to IgE EMPD fragments with high specificity, with little, if any, antibodies directed against heterologous Th epitopes or optional heterologous spacers. In particular, polyclonal antibodies raised in vaccinated animals can bind with high specificity to the central region of the loop structure encompassing IgE EMPD, as shown in figure 9.
Method
The present disclosure also relates to methods for making and using IgE EMPD peptide immunogen constructs, compositions, and pharmaceutical compositions.
Method for producing IgE EMPD peptide immunogen constructs
IgE EMPD peptide immunogen constructs of the present disclosure may be prepared using chemical synthesis methods well known to those of ordinary skill (see, e.g., fields et al, chapter 3in Synthetic Peptides:A User's Guide,ed.Grant,W.H.Freeman&Co, new York, NY,1992, p.77). IgE EMPD peptide immunogen constructs can be synthesized using automated Me Li Fude (Merrifield) solid phase synthesis using side chain protected amino acids, chemically protecting α -NH2 with t-Boc or F-moc, for example, on the application biosystems peptide synthesizer model 430A or 431 (Applied Biosystems Peptide Synthesizer Model 430A or 431). Preparation of IgE EMPD peptide immunogen constructs comprising combinatorial library peptides of Th epitopes can be accomplished by providing a mixture of alternative amino acids for coupling at a given variable position.
After assembly of the desired IgE EMPD peptide immunogen construct is completed, the resin is treated according to standard procedures, the peptide is cleaved from the resin, and the functional groups on the amino acid side chains are cleaved. The free peptide can be purified by HPLC and used, for example, for amino acid analysis or sequencing to describe biochemical properties. Methods for purification and characterization of peptides are well known to those of ordinary skill in the art to which the present invention pertains.
The quality of the peptides produced by this chemical process can be controlled and determined, and as a result reproducibility, immunogenicity and yield of IgE EMPD peptide immunogen constructs can be ensured. A detailed description of the manufacture of IgE EMPD peptide immunogen constructs synthesized by solid phase peptide is shown in example 1.
It has been found that a range of structural variations that allows for the preservation of the desired immune activity is more inclusive than a range of structural variations that allows for the preservation of the specific pharmaceutical activity of a small molecule drug or the presence of the desired activity and undesired toxicity in macromolecules co-produced with drugs of biological origin. Thus, peptide analogs having similar chromatographic and immunological properties to the desired peptide, whether deliberately designed or produced as a mixture of missing sequence byproducts unavoidable due to synthetic process errors, generally have the same effect as purified desired peptide formulations. Mixtures of engineered and unexpected analogues are also effective as long as a stringent QC procedure is established to monitor the manufacturing process and product evaluation process, ensuring reproducibility and effectiveness of the end products using these peptides.
Recombinant DNA techniques including nucleic acid molecules, vectors, and/or host cells can also be utilized to prepare IgE EMPD peptide immunogen constructs. Thus, nucleic acid molecules encoding IgE EMPD peptide immunogen constructs and immunologically functional analogs thereof are also included in this disclosure as part of the invention. Similarly, vectors (including expression vectors) comprising nucleic acid molecules and host cells comprising vectors are also included in the present disclosure as part of the invention.
Various exemplary embodiments also include methods of making IgE EMPD peptide immunogen constructs and immunologically functional analogs thereof. For example, the method may comprise the step of culturing a host cell comprising an expression vector comprising a nucleic acid molecule encoding an IgE EMPD peptide immunogen construct and/or an immunologically functional analogue thereof under conditions that express the peptide and/or analogue. Longer synthetic peptide immunogens can be synthesized using well known recombinant DNA techniques. These techniques may be provided in well known standard manuals with detailed experimental plans. To construct a gene encoding the peptide of the invention, the amino acid sequence is reverse translated to obtain a nucleic acid sequence encoding the amino acid sequence, preferably using codons most appropriate for the organism in which the gene is to be expressed. Next, synthetic genes are typically made by synthesizing oligonucleotides that encode the peptide and any regulatory factors (if necessary). The synthetic gene is inserted into a suitable cloning vector and transfected into a host cell. The peptide is then expressed under appropriate conditions appropriate to the chosen expression system and host. The peptides were purified and characterized using standard methods.
b. Method for producing immunostimulatory complexes
Various exemplary embodiments also include methods of making an immunostimulatory complex comprising an IgE EMPD peptide immunogen construct and a CpG Oligodeoxynucleotide (ODN) molecule. The stabilized immunostimulatory complexes (ISCs) are derived from the cationic portion of the IgE EMPD peptide immunogen construct and the polyanionic CpG ODN molecules. Self-assembled systems are driven by electrostatic neutralization of charge. The stoichiometry of the molar charge ratio of the cationic portion to the anionic oligomer of the IgE EMPD peptide immunogen construct determines the extent of association. Non-covalent electrostatic binding of IgE EMPD peptide immunogen constructs to CpG ODN is a completely reproducible process. The peptide/CpG ODN immunostimulatory complex aggregates facilitate presentation to "professional" Antigen Presenting Cells (APCs) in the immune system, thus further enhancing the immunogenicity of the complex. Such composites can be easily characterized during manufacture to control quality. peptide/CpG ISC was well tolerated in vivo. This novel microparticle system comprising CpG ODN and IgE EMPD fragment-derived peptide immunogen constructs was designed to take advantage of the generalized B cell mitogenesis (mitogeneity) associated with CpG ODN use, but promote balanced Th-1/Th-2 type responses.
CpG ODN in the disclosed pharmaceutical compositions bind 100% to the immunogen during the process mediated by oppositely charged electrostatic neutralization, resulting in the formation of micron-sized microparticles. The particulate form allows for a significant reduction in CpG doses from conventional use of CpG adjuvants, lower likelihood of adverse innate immune responses, and promotes alternative immunogenic processing pathways including Antigen Presenting Cells (APCs). Thus, such dosage forms are conceptually novel and offer potential advantages by promoting stimulation of immune responses through alternative mechanisms.
c. Method for producing pharmaceutical composition
Various exemplary embodiments also include pharmaceutical compositions comprising IgE EMPD peptide immunogen constructs. In certain embodiments, the pharmaceutical composition is in a dosage form utilizing a water-in-oil emulsion and a suspension with mineral salts.
In order to make pharmaceutical compositions available to a wide population and to avoid IgE EMPD aggregation also becomes part of the target of administration, safety is another important consideration. Although water-in-oil emulsions are used in humans for many dosage forms in clinical trials, alum is still the primary adjuvant used in the formulation based on its safety. Thus, alum or its mineral salt aluminum phosphate (ADJUPHOS) is often used as an adjuvant in formulations for clinical use.
Other adjuvants and immunostimulants include 3 De-O-acylated monophosphoryl lipid A (MPL) or 3-DMP, polymeric or monomeric amino acids, such as polyglutamic acid or polylysine. Such adjuvants may be used with or without other specific immunostimulants such as muramyl peptides (e.g., N-acetylmuramyl-L-threonyl-D-isoglutamyl (thr-MDP), N-acetyl-N-muramyl-L-alanyl-D-isoglutamyl (nor-MDP), N-acetylmuramyl-L-alanyl-D-isoglutamyl-L-alanyl-2- (1 '-2' -dipalmitoyl-sn-propyltri-3-hydroxyphosphoryloxy) -ethylamine (MTP-PE), N-acetylglucosamine-N-acetylmuramyl-L-A1-D-isoglu-L-Ala-dipalmitoylalanyl alanyl (DTP-DPP) Theramide TM ) Or other bacterial cell wall components. The oil-in-water emulsion comprises MF59 (see patent application WO 90/14837 to Van Nest et al,incorporated herein by reference in its entirety), comprising 5% squalene, 0.5% tween 80, and 0.5% span 85 (optionally containing different amounts of MTP-PE), formulated into sub-micron particles using a microfluidizer; SAF comprising 10% squalene, 0.4% Tween 80, 5% pluronic-block copolymer L121, and thr-MDP, forming a sub-micron emulsion by microfluidization or generating a large particle emulsion by vortex shaking; ribi TM An adjuvant system (RAS) (Ribi ImmunoChem, hamilton, mont.) comprising 2% squalene, 0.2% Tween 80, and one or more bacterial cell wall components selected from the group consisting of monophosphoryl lipid A (MPL), trehalose Dimycolate (TDM), and Cell Wall Skeleton (CWS), preferably MPL+CWS (Detox) TM ). Other adjuvants include Freund's complete adjuvant (CFA), freund's incomplete adjuvant (IFA), and cytokines such as interleukins (IL-1, IL-2 and IL-12), macrophage colony stimulating factor (M-CSF), and Tumor Necrosis Factor (TNF).
The choice of adjuvant depends on the stability of the immunogenic formulation containing the adjuvant, the route of administration, the dosing schedule, the efficacy of the adjuvant against the species of the vaccinated vaccine, and in humans refers to pharmaceutically acceptable adjuvants that have been approved or can be approved by the relevant regulatory authorities for human administration. Such as alum alone, MPL, or freund's incomplete adjuvant (Chang et al Advanced Drug Delivery Reviews 32:173-186 (1998), which is incorporated herein by reference in its entirety) or optionally all combinations thereof, are suitable for human administration.
The composition may include a pharmaceutically acceptable non-toxic carrier or diluent, which is defined as a carrier commonly used to formulate pharmaceutical compositions for administration to animals or humans. The diluent is selected so as not to affect the biological activity of the composition. Examples of such diluents are distilled water, physiological phosphate buffered saline, ringer's solution, dextrose solution, and Hank's solution. In addition, the pharmaceutical compositions or dosage forms may also contain other carriers, adjuvants or nontoxic, non-therapeutic, non-immunogenic stabilizers and the like.
The pharmaceutical compositions may also include large slowly metabolized macromolecules such as proteins, polysaccharides (e.g., chitin), polylactic acid, polyglycolic acid, and copolymers (e.g., latex functionalized agarose (latex functionalized sepharose), agarose (agarsose), cellulose, etc.), polymeric amino acids, amino acid copolymers, and lipid aggregates (e.g., oil droplets or liposomes).
The pharmaceutical compositions of the present invention may further comprise a suitable delivery vehicle. Suitable delivery vehicles include, but are not limited to, viruses, bacteria, biodegradable microspheres, microparticles, nanoparticles, liposomes, collagen microspheres, and helices (cochleates).
d. Methods of using pharmaceutical compositions
The disclosure also includes methods of using pharmaceutical compositions comprising IgE EMPD peptide immunogen constructs.
In certain embodiments, pharmaceutical compositions comprising IgE EMPD peptide immunogen constructs may be used to treat and/or prevent immunoglobulin E (IgE) -mediated allergic diseases, including, but not limited to, allergies to drugs, foods and insects, allergic rhinitis (hay fever), atopic dermatitis, allergic asthma, conjunctivitis, eczema, urticaria (urticaria), and acute allergies.
In some embodiments, the method comprises administering to a host in need thereof a pharmaceutically effective dose of a pharmaceutical composition comprising an IgE EMPD peptide immunogen construct. In certain embodiments, the method comprises administering a pharmaceutically effective dose of a pharmaceutical composition comprising an IgE EMPD peptide immunogen construct to a warm-blooded animal (e.g., human, cynomolgus monkey, mouse) to elicit a highly specific antibody that is cross-reactive with the IgE EMPD 1-52 peptide (SEQ ID NO: 2), the IgE EMPD 1-67 protein (SEQ ID NO: 1), whether recombinant IgE EMPD-containing protein (e.g., purified from a stabilized Flp-In CHO cell line transfected with recombinant DNA encoding the Fc portion of human IgG1 and IgEEMPD (gamma 1-em 67) of human membrane-bound IgE) or on the cell membrane of a cell bearing membrane-bound IgE (e.g., using a recombinant DNA encoding mIgE. Fc L A Ramos cell line transfected with recombinant DNA of (a).
In certain embodiments, pharmaceutical compositions comprising IgE EMPD peptide immunogen constructs may be used to treat and/or prevent IgE-mediated diseases by eliciting antibodies against IgE EMPD. Such antibodies are capable of (a) binding to B cells expressing mIgE and inducing antibody-dependent cellular cytotoxicity (ADCC) and apoptosis; (b) results in vivo reduction of basal levels of IgE in blood; (c) Resulting in vivo reduction of antigen-specific IgE levels in blood; and (d) reducing or eliminating IgE-mediated allergic pathology in IgE-mediated allergic disease patients.
e. In vitro function detection and in vivo efficacy concept verification test
Antibodies generated using IgE EMPD peptide immunogen constructs may be used for in vitro functional assays. These functional assays include, but are not limited to:
(a) In vitro binding to IgE EMPD 1-52 peptide (SEQ ID NO: 1) as a polypeptide derived from expression of mIgE. Fc L CHO cell purified recombinant protein of (example 3);
(b) In vitro binding to cells with membrane-bound IgE (Ramos) from B cell lines using IgE.Fc encoding L Recombinant DNA transfection of (example 3);
(c) In vitro Antibody Dependent Cellular Cytotoxicity (ADCC) (example 6);
(d) Apoptosis of IgE-bearing B lymphocytes is induced in vitro (example 6);
(e) Efficacy was demonstrated in vivo by showing a decrease in basal levels of IgE in the blood of immunized hosts (examples 8-10);
(f) Toxicity challenge by allergen after primary and booster immunization showed a decrease in antigen-specific IgE levels to verify efficacy in vivo (examples 8 to 10);
with the present disclosure, igE EMPD peptide immunogen constructs and dosage forms thereof may be effective as vaccines to reduce or eliminate IgE-mediated allergic pathologies in IgE-mediated allergic disease patients.
Detailed description of the preferred embodiments
(1) An IgE EMPD peptide immunogen construct represented by the formula:
(Th) m -(A) n - (IgE EMPD fragment) -X
Or (b)
(IgE EMPD fragment) - (A) n -(Th) m -X
Or (b)
(Th) m -(A) n - (IgE EMPD fragment) - (a) n -(Th) m -X
Wherein the method comprises the steps of
Th is a heterologous T helper epitope;
a is a heterologous spacer;
(IgE EMPD fragment) is a B cell epitope of about 20 to about 40 amino acid residues from the central intramolecular loop structure of IgE EMPD;
x is alpha-COOH or alpha-CONH of amino acid 2
m is 1 to about 4; and
n is 0 to about 10.
(2) The IgE EMPD peptide immunogen construct according to (1), wherein the IgE EMPD fragment is selected from the group consisting of SEQ ID NO: 5. 6, 8 and 9.
(3) The IgE EMPD peptide immunogen construct according to any one of (1) or (2), wherein the T helper epitope is selected from the group consisting of SEQ ID NO: 59-87.
(4) The IgE EMPD peptide immunogen construct according to (1), wherein the peptide immunogen construct is selected from the group consisting of SEQ ID NOs: 88-95, 98-124 and 130.
(5) An IgE EMPD peptide immunogen construct comprising:
comprising a sequence derived from SEQ ID NO:1 or SEQ ID NO:2 from about 20 to about 40 amino acid residues;
comprising a sequence selected from the group consisting of SEQ ID NOs: 59-87; and
An optional heterologous spacer selected from the group consisting of amino acids, lys-, gly-, lys-Lys-Lys-, (α, ε -N) Lys and ε -N-Lys-Lys-Lys (SEQ ID NO: 129);
wherein the B cell epitope is covalently linked to the T helper cell epitope either directly or through an optional heterologous spacer.
(6) The IgE EMPD peptide immunogen construct of (5), wherein the B cell epitope is selected from the group consisting of SEQ ID NO: 5. 6, 8 and 9.
(7) The IgE EMPD peptide immunogen construct of (5), wherein the T helper epitope is selected from the group consisting of SEQ ID NO: 59-87.
(8) The IgE EMPD peptide immunogen construct of (5) wherein the optional heterologous spacer is (. Alpha.,. Epsilon. -N) Lys or. Epsilon. -N-Lys-Lys-Lys-Lys (SEQ ID NO: 129).
(9) The IgE EMPD peptide immunogen construct of (5), wherein the T helper epitope is covalently linked to the amino terminus of the B cell epitope.
(10) The IgE EMPD peptide immunogen construct of (5), wherein the T helper epitope is covalently linked to the amino terminus of the B cell epitope through an optional heterologous spacer.
(11) A composition comprising a peptide immunogen construct according to any one of (1) to (10).
(12) A pharmaceutical composition comprising:
a. the peptide immunogen construct according to any one of (1) to (10); and
b. Pharmaceutically acceptable delivery vehicles and/or adjuvants.
(13) The pharmaceutical composition of (12), wherein
Ige EMPD peptide immunogen construct selected from the group consisting of SEQ ID NO:88-95, 98-124, and 130; and
ige EMPD peptide immunogen constructs were mixed with CpG Oligodeoxynucleotides (ODNs) to form stabilized immunostimulatory complexes.
(14) An isolated antibody or epitope-binding fragment thereof that specifically binds to a B cell epitope of an IgE EMPD peptide immunogen construct according to any one of (1) to (10).
(15) The isolated antibody or epitope-binding fragment thereof according to (14) binds to an IgE EMPD peptide immunogen construct.
(16) An isolated antibody or epitope-binding fragment thereof that specifically binds to a B cell epitope of an IgE EMPD peptide immunogen construct according to any one of (1) to (10).
(17) A composition comprising an isolated antibody or epitope-binding fragment thereof according to any one of (14) to (16).
The following examples provide detailed illustrations of the procedures used. The following examples are intended to illustrate the invention and are not intended to limit the scope of the invention.
EXAMPLE 1 Synthesis of IGE EMPD-related peptides and preparation of dosage forms thereof
Synthesis of IgE EMPD-related peptides
Methods for synthesizing specifically designed IgE EMPD-related peptides contained in IgE EMPD peptide immunogen construct development programs are described. Peptides synthesized in small scale amounts are used for serological analysis, laboratory tests and field trials, while peptides synthesized in large scale (kilogram) amounts are used for industrial/commercial production of pharmaceutical compositions. In order to screen and select the best peptide constructs for an effective IgE allergy-based vaccine, a number of IgE EMPD-related antigenic peptides were designed with sequences of about 20 to 70 amino acids in length.
As shown in Table 1 (SEQ ID NO:1 through 58), representative full-length IgE EMPD1-67 (SEQ ID NO: 1), igE EMPD 1-52 (SEQ ID NO: 2), and IgE EMPD fragments, including IgE EMPD1-17 (SEQ ID NO: 7), igE EMPD 19-38 (SEQ ID NO: 8), and various 10-mer peptides (SEQ ID NO: 10-58), were used for epitope identification in various serological assays.
Selected IgE EMPD B cell epitope peptides were made into IgE EMPD peptide immunogen constructs by synthetically ligating carefully designed T helper cell (Th) epitopes derived from pathogen proteins including measles virus fusion protein (MVF), hepatitis B surface antigen protein (HBsAg), influenza virus, clostridium tetani, and Epstein-Barr virus (EBV), as shown in Table 2 (SEQ ID NO: 59-87). Th epitopes are used in the form of single sequences (SEQ ID NOS: 59-67 and 72-87) or combinatorial libraries (SEQ ID NOS: 68-71) to enhance the immunogenicity of their respective IgE EMPD peptide immunogen constructs.
Representative IgE EMP D peptide immunogen constructs selected from more than 100 peptide constructs are identified in Table 3 (SEQ ID NOS: 88-124 and 130).
All peptides used for immunogenicity studies or related serological tests for detection and/or measurement of anti-IgE EMPD antibodies were synthesized on a small scale using F-moc chemistry on the application biosystems peptide synthesizers 430A, 431 and/or 433. Each peptide was prepared by independent synthesis on a solid support, with F-moc protection at the amino terminus of the trifunctional amino acid with the side chain protecting group. The complete peptide was cleaved from the solid support and the side chain protecting group was removed with 90% trifluoroacetic acid (TFA). The synthesized peptides were evaluated using a matrix assisted laser desorption free time of flight mass spectrometer (MALDI-TOF) to determine the correct amino acid composition. The synthetic peptides were evaluated by reverse phase HPLC (RP-HPLC) to confirm the synthetic profile and concentration of the product. Despite the tight control of the synthesis process (including stepwise monitoring of coupling efficiency), peptide analogs may still be produced due to certain unexpected events in the extended cycle, including insertion, deletion, substitution, and premature termination of amino acids. Thus, the synthesis product generally includes a variety of peptide analogs with the target peptide.
Despite the inclusion of these unexpected peptide analogs, the final synthetic peptide products are useful for immunological applications, including immunodiagnosis (as antibody capture antigens) and pharmaceutical compositions (as peptide immunogens). Generally, such peptide analogs, including mixtures of by-products generated during deliberate design or synthetic procedures, are generally as effective as the purified product of the desired peptide, provided that stringent QC procedures are developed to monitor the manufacturing process and product quality assessment procedures to ensure reproducibility and effectiveness of the final product using the peptides. A large amount of peptides of hundreds to thousands of grams can be synthesized on a scale of 15mmole to 50mmole using a custom-made automated peptide synthesizer UBI2003 or similar models.
For the active ingredient to be used in the final pharmaceutical composition for clinical trials, igE EMPD-related peptide constructs can be purified by preparative RP-HPLC under a shallow elution gradient, amino acid composition determined by MALDI-TOF mass spectrometry, and purity and consistency assessed by amino acid analysis and RP-HPLC.
b. Preparation of compositions comprising IgE EMPD peptide immunogen constructs
Dosage forms were prepared using water-in-oil emulsions and suspensions with mineral salts. In order to design pharmaceutical compositions for use in a broad population and to prevent them from also becoming part of the target of administration, safety is another important consideration. Although water-in-oil emulsions are used in clinical trials of many pharmaceutical compositions in humans, alum is still the primary adjuvant used in pharmaceutical compositions based on its safety. Thus, alum or its mineral salt, ADJUPHOS (aluminum phosphate), is often used as an adjuvant for clinical applications.
Briefly, the dosage forms specified in each of the experimental groups described below generally comprise all types of specially designed IgE EMPD peptide immunogen constructs. More than 100 specially designed IgE EMPD peptide immunogen constructs were initially evaluated in guinea pigs for their relative immunogenicity using corresponding IgE EMPD peptides representing the immunogenic B cell epitope peptides, and also using coating with a peptide selected from the group consisting of SEQ ID NOs: 1-126 in a well plate of different peptides, the serological cross-reaction between the various homologous peptides was evaluated in an ELISA assay.
(i) Using an approved oil for human use, seppic Montanide TM ISA 51 formulated as a water-in-oil emulsion, or (ii) mixed with mineral salts ADJUPHOS (aluminum phosphate) or aldrogel (alum) to formulate IgE EMPD peptide immunogen constructs, the peptide constructs being formulated in varying amounts as specified. Typically by dissolving IgE EMPD peptide immunogen constructs in water at a concentration of about 20 to 800 μg/mL and combining with Montanide TM ISA 51 is formulated as a water-in-oil emulsion (1:1 volume) or as a composition with mineral salts or ALHYDROGEL (alum) (1:1 volume). The composition was left at room temperature for about 30 minutes and mixed with vortex shaking for about 10 to 15 seconds prior to immunization. Animals were vaccinated with 2 to 3 doses of the particular composition, which were given by intramuscular route at time 0 (primary immunization) and 3 weeks after primary immunization (wpi) (boost), optionally 5 or 6wpi for a second boost. These immunized animals are then tested with the selected B cell epitope peptide to assess the immunogenicity of the various IgE EMPD peptide immunogen constructs present in the dosage form, as well as their cross-reactivity with the relevant peptide or protein of interest. Thereafter, those IgE EMPD peptide immunogen constructs that were effective in initial screening of guinea pigs were further tested in primates in water-in-oil emulsions, mineral salts and alum-based dosage forms for the dosing regimen within the specified period specified by the immunization protocol.
In the application of new drugs for testing and in the preparation of the submission of clinical trials in IgE-mediated disease patients, only the most promising IgE EMPD peptide immunogen constructs will be further extensively evaluated before being incorporated into the final dosage form for immunogenicity, duration, toxicity and efficacy studies in GLP-directed preclinical studies.
Example 2 serological assays and reagents
Serological assays and reagents for assessing the functional immunogenicity of synthetic peptide constructs and their dosage forms are described in detail below.
a. For antibody-specific analysisA kind of electronic deviceELISA assays based on IgE EMPD1-52, igE EMPD 1-39, igE EMPD 1-17, igE EMPD 19-38, igE EMPD7-40 peptides
ELISA assays for the evaluation of immune serum samples were developed and described in the examples below. The wells of 96-well plates were individually coated with target peptides IgE EMPD1-52, igE EMPD 1-39, igE EMPD 1-17, igE EMPD 19-38, igE EMPD7-40 peptides, etc. (e.g., SEQ ID NOs: 2 and 5 to 8) at a concentration of 2 μg/mL (unless otherwise indicated) in 10mM sodium bicarbonate buffer, pH9.5, at a volume of 100 μl for 1 hour at 37 ℃.
The peptide-coated wells were reacted with 250 μl gelatin in PBS at a concentration of 3% by weight for 1 hour at 37deg.C to block non-specific protein binding sites, followed by a reaction containing 0.05% by volume The wells were washed three times with 20 PBS and dried. With a mixture of 20 volume percent normal goat serum, 1 weight percent gelatin and 0.05 volume percent +.>20 in a 1:20 ratio (unless otherwise indicated) of PBS. mu.L of diluted sample (e.g. serum, plasma) was added to each well and reacted at 37℃for 60 minutes. Then using 0.05 volume percent of +.>The wells were washed 6 times 20 to remove unbound antibody. Horseradish peroxidase (HRP) -conjugated species (e.g., mouse, guinea pig, or human) -specific goat anti-IgG, igA, or IgM was used as a labeled tracer to bind to the formed antibody/peptide antigen complex in the positive well. 100 microliter of peroxidase-labeled goat anti-IgG was formulated at the optimal dilution of pre-titration to contain 1 volume percent normal goat serum and 0.05 volume percent20 in PBS, which was added to each well and reacted for an additional 30 minutes at 37 ℃. By inclusion of 0.05 volume percent->The wells were washed 6 times with 20 PBS to remove unbound antibody and reacted with 100 μl of a substrate mixture containing 0.04 weight percent 3',3',5',5' -Tetramethylbenzidine (TMB) and 0.12 volume percent hydrogen peroxide in sodium citrate buffer for an additional 15 minutes. The substrate mixture is used to detect peroxidase markers by forming colored products. The reaction was stopped by adding 100. Mu.L of 1.0M sulfuric acid and the absorbance at 450nm was determined (A 450 ). To determine antibody titers in vaccinated animals receiving various IgE EMPD peptide vaccine formulations, 10-fold serial dilutions of serum from 1:100 to 1:10,000 or 1:100 to 1:4.19x10 8 Is tested with 4-fold serial dilutions of serum and with A 450 A with a threshold of 0.5 450 The titer of the test serum was calculated by linear regression analysis of (C) in Log 10 And (3) representing.
b. Using Th-based peptidesA kind of electronic deviceELISA assay to assess antibody reactivity against Th peptides
Experiments were performed in a similar ELISA method as described above, using Th peptides formulated in 10mM sodium bicarbonate buffer at pH9.5 (unless otherwise indicated) at a concentration of 2. Mu.g/mL (unless otherwise indicated) and applied separately in a 100. Mu.L volume at 37℃for 1 hourWells of a 96-well ELISA plate were covered. To determine antibody titers in vaccinated animals receiving various IgE EMPD peptide vaccine formulations, 10-fold serial dilutions of serum from 1:100 to 1:10,000 were tested and used with A 450 A with a threshold of 0.5 450 The titer of the test serum was calculated by linear regression analysis of (C) in Log 10 The table is not shown.
c. Utilization of B cell epitope cluster-based 10-mer peptidesA kind of electronic deviceELISA assays Fine specificity analysis and epitope identification assessment against IgE EMPD and IgE CH4 (human IgE CH4 to cell membrane)
Fine specificity analysis of anti-IgE EMPD antibodies determined in immunized hosts using epitope identification. Briefly, wells of a 96-well plate were coated with 0.5. Mu.g of the individual IgE EMPD 10-mer peptides (SEQ ID NOS: 10 to 58) per 0.1mL of each well in a two-fold manner following the steps of the antibody ELISA method described above, and 100. Mu.L of serum samples (formulated in PBS at a dilution of 1:100) were then reacted in a 10-mer plate. For additional reactivity and specificity confirmation, B cell epitopes of IgE EMPD peptide immunogen constructs and anti-IgE EMPD antibody-related fine specificity assays from immune sera of immunized hosts were tested using the corresponding IgE EMPD 1-52 peptide (SEQ ID NO: 1) without spacers or Th sequences or an unrelated control peptide.
d. Immunogenicity assessment
Pre-immune and immune serum samples from animal or human subjects were collected following the experimental vaccination program and heated at 56 ℃ for 30 minutes to inactivate serum complement factors. After administration of the vaccine dosage form, blood samples are obtained according to the procedure and evaluated for immunogenicity against a specific target. Serial dilutions of serum were tested and the reciprocal of the dilution was Log (Log 10 ) To represent positive titers. The ability to elicit high titers of antibodies against the desired B cell epitope sequences within the antigen of interest is exploited to assess the immunogenicity of a particular vaccine dosage form while maintaining antibody reactivity against T helper cell epitope sequences to provide the desired B cell response as low as negligible.
e. Immunoassay for assessing human IgE levels in mouse serum
The human IgE levels in huige gene knockout mice were determined by sandwich ELISA using anti-human IgE, HP6061 (Abcam) as the capture antibody and biotin-labeled anti-human IgE, HP6029 (Abcam) as the detection antibody. Briefly, HP6061 was formulated in coating buffer (15 mM sodium carbonate, 35mM sodium bicarbonate, pH 9.6) at 100 ng/well to be immobilized on 96-well plates and reacted overnight at 4 ℃. The coated wells were blocked by reaction with 200. Mu.L/well assay dilution (PBS containing 0.5% BSA,0.05% Tween-20,0.02% liquid biological preservative ProClin 300) for 1 hour at room temperature. The microplate was washed 3 times with 200. Mu.L/well of wash buffer (PBS containing 0.05% Tween-20). Standard curves (ranging from 0 to 800ng/mL by 2-fold serial dilutions) were generated in assay dilutions containing 5% mouse serum using purified U266 IgE. 50. Mu.L of diluted serum (1:20) and standard were added to the coated wells. The reaction was carried out at room temperature for 1 hour. All wells were blotted dry and washed 6 times with 200 μl/well of wash buffer. The captured human IgE was reacted with 100 μl of detection antibody solution (50 ng/ml biotin-labeled HP6029 in assay dilution) at room temperature for 1 hour. Bound biotin-HP 6029 was then detected using streptavidin poly-HRP (1:10,000 dilution, thermo Pierce) for 1 hour (100. Mu.L/well). All wells were blotted dry and washed 6 times with 200 μl/well of wash buffer. Finally, the wells were developed using 100. Mu.L/well of NeA-Blue TMB substrate (Clinical Scientific Products) and the reaction was stopped by adding 100. Mu.L/well of 1M sulfuric acid. Standard curves were generated by 4-parameter logistic curve fitting using SoftMax Pro software (Molecular Devices) and used to calculate IgE concentrations in all test samples. Data were compared by Prism software using Student t-test.
f. Immunoassay for assessing papain-specific IgE levels in mouse serum
Papain-specific IgE formed in huige gene knock-in mice was determined by direct ELISA using papain as the coating material and biotin-labeled anti-human IgE, HP6029 (Abcam) as the detection antibody. Briefly, papain was formulated in coating buffer (15 mM sodium carbonate, 35mM sodium bicarbonate, pH 9.6) at 500 ng/well to be immobilized on 96-well plates and reacted overnight at 4 ℃. The coated wells were blocked by reaction with 200. Mu.L/well assay dilution (PBS containing 0.5% BSA,0.05% Tween-20,0.02% liquid biological preservative ProClin 300) for 1 hour at room temperature. The microplate was washed 3 times with 200. Mu.L/well of wash buffer (PBS containing 0.05% Tween-20). Standard curves (ranging from 0 to 30ng/mL by 2-fold serial dilutions) were generated in assay dilutions with 10% mouse serum using monoclonal human chimeric papain-specific IgE (aller mabs co., ltd.). 50. Mu.L of diluted serum (1:10) and standard were added to the coated wells. The reaction was carried out at room temperature for 1 hour. All wells were blotted dry and washed 6 times with 200 μl/well of wash buffer. The captured human IgE was reacted with 100 μl of detection antibody solution (50 ng/ml biotin-labeled HP6029 in assay dilution) at room temperature for 1 hour. Bound biotin-HP was then detected using streptavidin poly-HRP (1:10,000 dilution, thermo Pierce) for 60291 hours (100. Mu.L/well). All wells were blotted dry and washed 6 times with 200 μl/well of wash buffer. Finally, the wells were developed using 100. Mu.L/well of NeA-Blue TMB substrate (Clinical Scientific Products) and the reaction was stopped by adding 100. Mu.L/well of 1M sulfuric acid. Standard curves were generated by a 4-parameter logistic curve fit using SoftMax Pro software (Molecular Devices) and used to calculate papain-specific IgE concentrations in all test samples. Data were compared by Prism software using Student t-test.
g. Immunoassay to assess IgE levels in cynomolgus monkey serum
Cynomolgus IgE levels in cynomolgus monkeys were determined using a sandwich ELISA using anti-human IgE, MB10-5C4 (Miltenyi Biotec) as capture antibody and biotin-labeled polyclonal anti-cynomolgus IgE (Alpha Diagnostic International inc.) as detection antibody. Briefly, MB10-5C4 was formulated in a coating buffer (15 mM sodium carbonate, 35mM sodium bicarbonate, pH 9.6) at 100 ng/well to be immobilized on a 96-well plate and reacted overnight at 4 ℃. The coated wells were blocked by reaction with 200. Mu.L/well assay dilution (PBS containing 0.5% BSA,0.05% Tween-20,0.02% liquid biological preservative ProClin 300) for 1 hour at room temperature. The microplate was washed 3 times with 200. Mu.L/well of wash buffer (PBS containing 0.05% Tween-20). Standard curves (ranging from 0 to 10,000 ng/mL by 2-fold serial dilutions) were generated using purified cynomolgus IgE in assay dilutions with 10% cynomolgus serum. 100. Mu.L of diluted serum (1:10) and standard were added to the coated wells. The reaction was carried out at room temperature for 1 hour. All wells were blotted dry and washed 6 times with 200 μl/well of wash buffer. The captured human IgE was reacted with 100 μl of detection antibody solution (50 ng/ml biotin-labeled HP6029 in assay dilution) at room temperature for 1 hour. Bound biotin-HP was then detected using streptavidin poly-HRP (1:10,000 dilution, thermo Pierce) for 60291 hours (100. Mu.L/well). All wells were blotted dry and washed 6 times with 200 μl/well of wash buffer. Finally, the wells were developed using 100. Mu.L/well of NeA-Blue TMB substrate (Clinical Scientific Products) and the reaction was stopped by adding 100. Mu.L/well of 1M sulfuric acid. Standard curves were generated by a 4-parameter logistic curve fit using SoftMax Pro software (Molecular Devices) and used to calculate IgE concentrations in all test samples. Data were compared by Prism software using Student t-test.
EXAMPLE 3 evaluation of IgE EMPD peptide immunogen constructs and their formulation induced in animalsA kind of electronic deviceFunctional Properties of antibodies
Recombinant soluble IgE EMPD proteins and polypeptides encoding mIgE. Fc L (EMPD from CH2 to CM) or mIgE. Fc S Recombinant DNA transfected (CH 2 to CM does not contain EMPD) Ramos cell lines were tested for their ability to test immune serum or purified anti-IgE EMPD antibodies from the immunized host.
a. Cells
Ramos cell lines were purchased from American type culture Collection (ATCC, manassas, va.) and supplemented with 10% heat inactivation FBS (Invitrogen), 4mM L-glutamine, 25mM HEPES and1mM sodium pyruvate (Invitrogen; complete RPMI medium) in RPMI 1640 medium (Invitrogen, carlsbad, calif.). By encoding mIgE. Fc L Or mIgE. Fc S Is transfected into Ramos cells. Transfection of an mIgE.Fc with a DNA fragment encoding an isoform fragment of mIgE epsilon chain length (extending from CH2 to cytoplasmic peptide, containing EMPD) L Is a Ramos cell of (C). Transfection of an mIgE.Fc with a DNA fragment encoding a short or common isoform fragment of mIgE epsilon chain (extending from CH2 to cytoplasmic peptide, not containing EMPD) S Is a Ramos cell of (C). Will express mIgE. Fc L Or mIgE. Fc S Stable transfectants of Ramos cells were maintained in complete RMPI 1640 medium supplemented with 400mg/ml giemycin (Zeocin) (Invitrogen).
b. For ELISA testA kind of electronic devicePreparation of recombinant soluble IgE EMPD proteins
Flp-In CHO cells expressing recombinant soluble IgE EMPD proteins were transfected with a DNA fragment encoding the Fc portion of human IgG1 and IgE EMPD (γ1-em 67) of human membrane-bound IgE. Stable Flp-In CHO transfectants were maintained In IMDM medium (Invitrogen, carlsbad, calif.) supplemented with 10% heat-inactivated FBS (Invitrogen), 4mM L-glutamine, 25mM HEPES and 1mM sodium pyruvate (Invitrogen; complete IMDM medium). The gamma 1-em67 protein was purified from the medium using protein A Sepharose (GE Healthcare) according to the manufacturer's instructions.
c. Purification of polyclonal antibodies from immune serum
Polyclonal IgG from various immune sera was purified using protein a Sepharose (GE Healthcare) according to the manufacturer's instructions.
d. Binding to soluble IgE EMPD proteins or mIgE.Fc located on B cells using polyclonal antibodies L
For its binding to (a) recombinant γ1-em67 protein (as described above) (using ELISA), or (b) expression of mIgE. Fc L The relative activity of Ramos cells (analyzed by fluorescence flow cytometry) from each immunized animal was examined for purified polyclonal antibodies. 96 well microplates were from Nalge NUNC International, flat bottom (accession No. 442404) for optical reading and V bottom (accession No. 249570) for cell culture. In Versamax micro The optical density is read on a disk analyzer (Molecular Devices). Fluorescence staining was detected using a BD FACSCanto II flow cytometer (DB Biosciences); and obtain the result data using the federated FACSDiva software. Binding data from ELISA and FACS were entered into Prism 6 software for quantitative analysis. More specifically, the microplate addition at V-bottom has 2X10 in 0.1mL per well 5 A solution of individual cells, which are centrifuged and the liquid removed. Cells were incubated on ice for 1 hour with solutions of different concentrations of 100 μl antibody samples. Cells were washed once, centrifuged at 300g for 5 min, and 100. Mu.L goat F (ab) was used 2 Anti-species specific IgG Fc-FITC (250 ng/mL) was stained on ice for 30 min. Cells were washed once and after centrifugation the liquid was removed. 200. Mu.L of binding buffer was added to each well and transferred to a microtiter plate for flow cytometry analysis. Binding intensities (geometric mean of fluorescence intensities, geoMFI) based on 10,000 cell injections per sample were read on FACS.
e. Apoptosis assay
Will stably express mIgE. Fc L Ramos cells (5 x 10) 5 cells/mL) were incubated with purified immune or control antibodies in complete RPMI 1640 medium for 1 hour at 37 ℃. Goat F (ab') specific for Fc of guinea pig IgG was then raised against a final concentration of 10. Mu.g/mL using a secondary antibody 2 (Jackson ImmunoResearch Laboratories, west Grove, pa.) cells were treated and incubated at 37℃for an additional 24 hours. The degree of apoptosis of the cells was analyzed in the following manner. For assays using annexin V, the exposure of Phosphatidylserine (PS) was determined by re-suspending cells in staining solution for 15 minutes at room temperature in the dark. The staining solution contained FITC-labeled annexin V (Biovision, mountain View, calif.) and 2.5 μg/ml Propidium Iodide (PI) diluted 1/200 in a buffer with 10mM HEPES/sodium hydroxide (pH 7.4), 140mM sodium chloride and 5mM calcium chloride. Stained cells were analyzed on a FACSCanto II flow cytometer (BD Biosciences, san Jose, CA). The percentage of apoptotic cells, defined as annexin V positive and PI negative, was obtained in the dot plot analysis.
f. Antibody Dependent Cellular Cytotoxicity (ADCC) assays
Spleen lymphocytes were isolated from the spleen of Balb/c mice (females, 6 to 8 weeks old) by repeated hypoosmotic shock reactions of erythrocytes using RBC lysis buffer (Thermo Fisher Scientific inc.). After removal of erythrocytes, the cells were removed in complete RPMI medium supplemented with 50. Mu.M 2-ME and 100U/mL recombinant human IL-2 (PeproTech, inc.) at 3X10 6 Cell concentration of cells/mL spleen lymphocytes were cultured for 3 days. Labeling of expression of mIgE. Fc in PBS/0.1% BSA at 37℃with CFSE (Invitrogen) L Ramos cells (target cells) of (E) for 10 minutes. After three washes with cold complete RPMI1640 medium, the cells were adjusted to 10 5 cells/mL. A solution containing 20,000 labeled cells in 200. Mu.l of complete RPMI medium was coated with purified polyclonal IgG antibodies (concentration 10. Mu.g/mL) from the corresponding immune serum at 37℃for 30 minutes, and then combined with IL-2 activated spleen lymphocytes (effector cells) at an E/T ratio of 30. After 24 hours of incubation, total cells were stained with 7-amino actinomycin D (7-AAD, invitrogen) at a concentration of 2.5. Mu.g/mL for 15 minutes on ice and then analyzed on a Becton Dickinson FACS Canto II flow cytometer (BD Biosciences). Live target cells were defined as CFSE positive and 7-AAD negative in the dot pattern analysis. The percentage of lysed target cells at a given E/T ratio is: 100x [ (percentage of live target cells in antibody-independent control-percentage of live target cells in sample)/percentage of live target cells in antibody-independent control ]。
EXAMPLE 4 animals for safety, immunogenicity, toxicity and efficacy studies
a. Guinea pigs:
at maturity, either without contact with or stimulation by antigenImmunogenicity studies were performed in adult male and female Duncan-Hartley guinea pigs (300-350 g/BW). At least 3 guinea pigs were used for each group in the experiment. In contracted animal facilities and in combined biomedical companies as test entrustersA test plan involving Duncan-Hartley guinea pigs (8-12 span; covance Research Laboratories, denver, pa., USA) was performed in accordance with the approved IACUC application.
b. Cynomolgus monkey:
immunogenicity and repeat dose toxicity studies were performed in adult male and female monkeys (cynomolgus monkeys, about 4 years; joinn Laboratories, suzhou, china) in contracted animal facilities and UBI as test taker in accordance with the approved IACUC application.
hIGHE gene knock-in mice:
the mouse strain was made to express human secreted and membrane bound IgE (Lu, el al, 2015) using homologous genes targeting the C57BL/B6 genetic background, replacing its IGHG1 gene with the human IGHE gene. The hIGHE mice express human IgE under the control of a murine IGHG1 transcriptional module and express human membrane-bound IgE by alternative RNA splicing of the human regulatory module. Serum IgE was detected in the circulation as early as 8 to 10 weeks of age. Young offspring (10-12 weeks old) of mixed hIGHE x Balb/c genetic background were used in a primary/memory immune response (primary/memory immune response) preventative model, and in a sensitization/recall immune response (sensitization/recall immune response) treatment model. Both studies were conducted at contracted animal facilities (National Health Research Institute, taiwan) and UBI as a test commissioner in accordance with the approved IACUC application.
The effect of intramuscular vaccination over a 16 week period was observed using ELISA analysis of serum human IgE for antibody responses, as well as evidence of reduced levels of antigen-specific IgE following challenge with serum total IgE and antigen.
Prior to immunization, serum samples from individual animals were tested for the presence of serum human IgE according to the method described above in this example. Depending on the species and the test plan, each animal was immunized with each dose of IgE EMPD peptide immunogen construct of the vaccine dosage form.
Example 5 selection of vaccine dosage forms for immunogenicity assessment of IgE EMPD peptide constructs in guinea pigs, transgenic knock-in mice and cynomolgus monkeys for final products
The pharmaceutical compositions and vaccine dosage forms used in each experiment are described in more detail below. Briefly, the dosage forms specified in each test group typically comprise all types of specially designed IgE EMPD peptide immunogen constructs having variants of IgE EMPD peptide fragments and promiscuous T helper cell epitopes (comprising two sets of artificial T helper cell epitopes derived from measles virus fusion proteins and hepatitis B surface antigen) linked via different kinds of spacers (e.g. epsilonks or KKKs to enhance the solubility of the peptide construct) linked to the amino terminus of the specially designed peptide construct. Over 100 specially designed IgE EMPD peptide constructs were initially evaluated in guinea pigs for their relative immunogenicity to IgE EMPD 1-52, and further for their cross-reactivity with cell membrane IgE located on IgE-bearing B cells (Ramos cell lines). As indicated, in the case of different amounts of peptide construct, seppic Montanide was used TM ISA 51 as an approved oil for use in human vaccines either formulated IgE EMPD peptide constructs in water-in-oil emulsions or mixed with mineral salts ADJUPHOS or aldrogel (alum). Vaccine preparation typically utilizes Montanide by dissolving the IgE EMPD peptide construct in water at a concentration of about 20 to 800 μg/mL TM ISA 51 is formulated as a water-in-oil emulsion (1:1 volume) or with the mineral salts ADJUPHOS or alum (1:1 volume). The vaccine dosage form was kept at room temperature for about 30 minutes and mixed with vortex shaking for about 10 to 15 seconds prior to immunization.
Animals were immunized with 2 to 3 doses of the particular vaccine dosage form, which were given by intramuscular route at time 0 (primary immunization) and 3 weeks after primary immunization (wpi) (booster immunization), optionally with 5 or 6wpi for a second booster immunization. These immunized animals were then tested to assess the immunogenicity of the various synthetic IgE EMPD peptide immunogens present in the vaccine dosage form, as well as their cross-reactivity with IgE EMPD 1-52. Thereafter, igE EMPD peptide immunogens with potent immunogenicity in guinea pig initial screening were further tested in cynomolgus monkeys in water-in-oil emulsions, mineral salts and alum-based dosage forms for dosing regimens within the specified period of time specified by the immunization protocol.
In the preparation of new drug applications for testing and the submission of clinical trials in IgE-mediated allergic disease patients, only the most promising IgE EMPD peptide immunogenic candidates would be further evaluated by the immune serum capacity mediating ADCC, apoptosis of B cells bearing mIgE, and in transgenic knock-in mice and cynomolgus monkeys, against their ability to break immune tolerance in the same species with autologous IgE EMPD antigen, before being included in the final vaccine formulation for GLP-directed immunogenicity, duration, toxicity and efficacy validation studies.
Example 6 design principles, screening, identification, functional Property assessment and optimization of multicomponent vaccine dosage forms incorporating IgE EMPD 1-39 peptide immunogen constructs for IgE mediated allergic disease treatment
FIGS. 2A and 2B illustrate the rationale for depletion of mIgE B cells by targeting IgE EMPD. IgE is expressed in two forms: secreted IgE and membrane-bound IgE (mIgE) (left and right side of fig. 2A, respectively). Secreted IgE is captured on the cell surface of basophils and mast cells by fceri, whereas mIgE is present only on IgE-directed B cells as part of the B Cell Receptor (BCR). The outer membrane proximal domain (EMPD) of mIgE is a 67 amino acid peptide fragment (SEQ ID NO: 1) located between the CH4 domain and the transmembrane region found on mIgE B cells only. The uniqueness of IgE EMPD provides an attractive site for targeting B cells with mIgE. Depletion of mIgE B cells by targeting IgE EMPD allowed inhibition of allergen-specific IgE production before it differentiated into new IgE-secreting plasma cells (fig. 2B). Existing IgE-secreting plasma cells with limited life time gradually die out, resulting in a gradual decrease in total IgE and allergen-specific IgE.
Fig. 3 is a flow chart identifying the development of vaccine dosage forms from discovery to commercialization (industrialization) according to certain embodiments disclosed herein. As summarized in this manner, the present disclosure includes peptide immunogen design, peptide composition design, vaccine dosage form design, in vitro functional antigenicity design, in vivo immunogenicity and efficacy study design, and clinical trial planning design. The detailed evaluation for each step surprisingly leads to a series of experiments leading to the final commercialization of safe and effective vaccine dosage forms.
The following describes a generalized summary of these steps:
a. design history
Each peptide immunogen construct or immunotherapeutic product requires its own design emphasis and methodology based on its specific disease mechanism and the target protein required for intervention. The goal of the model design may then include cellular proteins involved in the disease pathway or infectious agents in which multiple proteins from pathogens may be involved. The process from research to commercialization is very long and typically requires one or more decades to complete.
Once the target molecule is selected, extensive serological validation procedures are required. Identification of B-cell and T-cell epitopes and functional sites located within the target molecule in accordance with the intervention is important for the design of the immunogenic construct. Once the target B cell epitope is recognized, continuous pilot immunogenicity studies incorporating various T helper supports (carrier proteins or suitable T helper peptides) into small animals are performed to assess the functional properties of antibodies raised using pharmaceutical compositions of specifically designed peptides. This serological application was then performed in animals of the target species to further verify immunogenicity and functional properties of the raised antibodies. Evaluation was performed using serum collected from immunized hosts, all studies were performed in multiple parallel groups. For human pharmaceutical compositions, early immunogenicity studies in the target species or non-human primate are also performed to further verify the immunogenicity and direction of the design. Then, when used in combination to prepare individual dosage form designs, the peptides of interest are formulated in different mixtures to assess subtle differences in functional properties associated with the respective interactions between the peptide constructs. After additional evaluation, the final peptide construct, peptide composition and dosage form thereof, as well as the respective physical parameters of the dosage form, are established in this way leading to the final product development process.
b. Design and validation of IgE EMPD-derived peptide immunogen constructs as pharmaceutical compositions for patients with potential treatment of IgE-mediated allergic diseases
To generate the most effective peptide constructs for inclusion in pharmaceutical compositions, a number of IgE EMPD B cell epitope peptides (SEQ ID NOS: 5-8) (Table 1) and promiscuous T helper cell epitopes derived from various pathogens or further engineered artificial T helper cell epitopes (SEQ ID NOS: 59-87) (Table 2) were made into IgE EMPD peptide immunogen constructs for guinea pig immunogenicity studies.
i) IgE EMPD G1-H40 was selected as the target region for immunogen design
IgE is expressed in two forms: secreted IgE and membrane-bound IgE (mIgE). Secreted IgE is captured on the cell surface of basophils and mast cells by fceri, whereas mIgE is present only on IgE-directed B cells as part of the B Cell Receptor (BCR). The full-length Extracellular Membrane Proximal Domain (EMPD) of mIgE is a 67 amino acid peptide fragment (SEQ ID NO: 1) located between the CH4 domain and the transmembrane region found on mIgE B cells only. Existing IgE-secreting plasma cells with limited life time gradually die out, resulting in a gradual decrease in total IgE and allergen-specific IgE. Many peptide immunogen constructs were tested against immunogenicity in guinea pigs, data from a series of IgE EMPD-derived peptide immunogen constructs (SEQ ID NO: 88-93) combining a representative JgE EMPD B cell epitope peptide derived from IgE EMPD 1-52 (SEQ ID NO: 2) with a representative Th epitope peptide (SEQ ID NO: 72) provides the use of IgE EMPD 1-39 (SEQ ID NO: 5) peptides to coat microplates in guinea pigs for testing immunogenicity as shown in Table 4. It was found that six IgE EMPD peptide immunogen constructs with three orientations were highly immunogenic, wherein the spacer linker using εK wouldThe Th epitope peptide is linked to the IgE EMPD B cell epitope peptide, which is linked to the carboxy-terminus (SEQ ID NO:88 and 91) or the amino-terminus (SEQ ID NO:89, 90 and 92) or both the carboxy-terminus and the amino-terminus (SEQ ID NO: 93) of the IgE EMPD B cell epitope peptide. Comprising longer linkers εK-IgE EMPD peptide immunogen constructs of KKK (SEQ ID NO: 129) have also been used to achieve high immunogenicity in the construction design (e.g., SEQ ID NO: 94-97).
Shorter fragments of IgE EMPD B cell epitope peptides (SEQ ID NOS: 7 and 8) may also be used with T helper cell epitope peptides (e.g) Enhancing immunogenicity (SEQ ID NO:96 and 97). However, as shown in Table 5, these constructs elicit antibodies with weak binding capacity to IgE EMPD (e.g., for longer fragments with SEQ ID NOs: 5, 6, 7 and 8). SEQ ID NO:96 and 97 may also elicit antibodies with very restricted binding patterns as shown in linear epitope identification studies, where antibodies with SEQ ID NOs: 25 (amino acids 8-17) and SEQ ID NO:39 Only the 10mer peptides of (amino acids 22-31) are reactive. Furthermore, SEQ ID NO:96 and 97 have minimal mIgE-B cell binding effects (as shown in figure 6C), a requirement for induction of mIgE-B cell apoptosis.
ii) heterologous T helper cell epitopes derived from pathogens designed to enhance the immunogenicity of selected IgE EMPD B cell epitope peptides and ordering of their contents in IgE EMPD peptide immunogen constructs
Table 2 lists a total of 29 heterologous Th epitopes (SEQ ID NO: 59-87) that have been tested for their relative efficacy in enhancing B cell epitope immunogenicity in multiple species of mice, rats, guinea pigs, baboons, cynomolgus monkeys, etc.
Representative experiments in guinea pigs with IgE EMPD peptide immunogen constructs containing IgE EMPD 1-39B cell epitope peptides (SEQ ID NO: 5) linked by epsilon K spacers to individual promiscuous T helper cell epitopes (SEQ ID NO:88, 98-124 and 130) were performed for immunogenicity studies to rank the relative potency of the respective heterologous T helper cell epitopes as shown in Table 6. Since some Th epitopes have high B cell epitope enhancing ability, the results obtained 3 weeks after the initial immunization (3 wpi) after only one immunization of guinea pigs were used to rank the 29 different IgE EMPD peptide immunogen constructs. Although all selected Th epitopes have the ability to enhance the immunogenicity of IgE EMPD B cell epitope peptides, the most effective construct was found to be SEQ ID NO:88, while the least potent is the construct of SEQ ID NO: 101.
Careful verification of the immunogenicity of each and all IgE EMPD peptide immunogen constructs in different species, including primates, will ensure final Th peptide selection and successful development of the final vaccine formulation.
iii) Immunogenicity assessment of IgE EMPD G1-C39 peptide immunogen constructs (antibody reactivity against the corresponding full-length and IgE EMPD G1-C39 peptides)
FIG. 4 illustrates the kinetics of antibody responses over a period of 8 weeks in guinea pigs immunized with different IgE EMPD peptide immunogen constructs (SEQ ID NOS: 88 to 94, 96 and 97). Serum was diluted from 1:100 to 1:10,000,000 with 10-fold serial dilutions. ELISA plates were coated with 0.5. Mu.g peptide IgE EMPD 1-39 peptide (SEQ ID NO: 5) per well. By A 450 A with a threshold of 0.5 450 The titer of the test serum was calculated by linear regression analysis of (C) in Log 10 And (3) representing.
FIG. 5 illustrates titration curves for various purified polyclonal anti-IgE EMPD antibodies generated using different IgE EMPD immunogen constructs (SEQ ID NOS: 88 to 94, 96 and 97). Using a polypeptide comprising SEQ ID NO:1 sequence containing IgE EMPD protein, gamma 1-em67, ELISA plate. Polyclonal anti-IgE EMPD antibodies purified from guinea pig serum by protein A chromatography were diluted from 100. Mu.g/mL to 0.0238ng/mL using 4-fold serial dilutions. EC for each polyclonal anti-IgE EMPD antibody preparation was calculated using nonlinear regression of 4-parameter logistic curve fit 50
Fig. 4 and 5 show the sequences from SEQ ID NO:88 to 94 are selected from a number of other designs, shown to have Log 10 The potency is mostly higher than 4. More accurate EC using purified antibodies from each group 50 The measurements are shown in FIG. 5, compared to peptide immunogen constructs containing IgE EMPD shorter B cell epitope peptides (length < 20 residues), such as IgE EMPD1-17 or IgE EMPD 19-38 (SEQ ID NO:96 and97 Using 0.02111 to 0.08892 μg/mL of IgE EMPD peptide immunogen construct comprising longer B-cell epitope peptides of IgE EMPD 1-39 and IgE EMPD 7-40 (SEQ ID NO: 88-94), shows a rather high immunogenicity.
IgE EMPD peptide immunogen constructs containing longer spacers located between B cells and Th epitopes also significantly reduced immunogenicity as in the case of IgE EMPD B cell epitope peptide design (SEQ ID NO:94vs 89 for the respective EC 50 0.08892vs 0.02368). Among the B cell epitope peptides of more than 20 residues in length, igE EMPD 1-39 was found to be optimal in design, and therefore will be most commonly used as a B cell epitope peptide in a representative peptide immunogen design in the following examples for further evaluation using various in vitro functional assays and in vivo efficacy assessments.
iv) immunogenicity assessment of IgE EMPD peptide immunogen constructs (binding of antibodies thereto to directed B cells differentiated into IgE secreting B cells bearing IgE)
FIGS. 6A through 6C illustrate the expression of mIgE.Fc using purified polyclonal antibodies from pooled guinea pig sera from each group of animals immunized with IgE EMPD immunogen constructs (SEQ ID NOS: 88 through 97) L Or mIgE. Fc S B cells derived from Ramos cell lines (see cell fluorescent staining methods of example 2). Polyclonal anti-IgE EMPD antibodies purified from guinea pig serum using protein A chromatography were used at a concentration of 10. Mu.g/mL.
The use of purified antibodies from all groups of high specificity from the cell binding assay found that the antibodies bound to a low background of cells from the mige.fcs Ramos cell line lacking the EMPD fragment of the target sequence. As shown in fig. 6A to 6C, the use of SEQ ID NO:88 to 94, 96 and 97 IgE EMPD peptide immunogen constructs L Ramos cell line cells have different binding affinities. Specifically, antibodies produced from IgE EMPD peptide immunogen constructs comprising the long B cell epitope peptide IgE EMPD 1-39 (SEQ ID NO: 88-94) showed mIgE.Fc compared to antibodies produced from IgE EMPD peptide immunogen constructs comprising B cell epitope peptides having less than 20 residues (SEQ ID NO: 96-97) L Ramos cell lineHigher ige binding of cells. Furthermore, antibodies produced from IgE EMPD peptide immunogen constructs with shorter spacers (εK) have mIgE. Fc compared to constructs with longer spacers (εK-KKK; SEQ ID NO: 129) L Higher mIgE binding by Ramos cell line cells, which can be seen from the sequence set forth in SEQ ID NO:89 and the sequence of SEQ ID NO:94, as can be seen by comparison of the results.
Discovery of polyclonal antibodies in group EC 50 The number was inversely related to the percent (%) positive binding with IgE cells. The percentage of IgE-bearing B-cell binding is an important functional parameter for assessing the cross-reactivity and functional efficacy of antibodies for the immunogenicity of IgE EMPD peptide immunogen constructs.
v) immunogenicity assessment of IgE EMPD peptide immunogen constructs (ability to produce antibodies directed against apoptosis of B cells with high potency to elicit differentiation of directed B cells into IgE-secreting B cells)
FIG. 7 shows the expression of mIgE.Fc using various formulations of polyclonal anti-IgE EMPD antibodies produced from IgE EMPD immunogen constructs (SEQ ID NOS: 88 to 93) L Apoptosis is triggered in a dose-dependent pattern on the cell surface of Ramos cells. Polyclonal anti-IgE EMPD antibodies purified from guinea pig serum by protein a chromatography were diluted from 1000 to 62.5ng/mL using 2-fold serial dilutions. Use of humanized anti-IgE monoclonal antibodies As a positive control. EC calculation of each group of polyclonal anti-IgE EMPD antibodies using nonlinear regression of 4-parameter logistic curve fit 50
As shown in fig. 7, although(anti-IgE monoclonal antibodies targeting the Fc receptor binding CH3 domain of IgE molecules) were shown to have the lowest EC indicating the highest efficacy 50 Numerical value (121.4 ng/mL), but its presence in serum neutralizes all such apoptosis-inducing capacity by being neutralized by serum IgE. After the selected IgE EMPD peptide immunogen construct tested (SEQ ID NO: 88-93), an EC of from 277.5 to 536ng/mL is observed 50 The values indicate the high ability to induce apoptosis without serum IgE interference due to their binding to unique EMPD target region sequences. These IgE-bearing B cells are cells that direct the differentiation of B cells into IgE secretion.
Immunization of a host with an IgE EMPD peptide immunogen construct (e.g., SEQ ID NO: 88-93) to induce apoptosis of these cells will trigger inhibition of IgE synthesis, resulting in serum loss of IgE, a major cause of allergic disease.
vi) immunogenicity assessment of IgE EMPD peptide immunogen constructs (ability to produce antibodies thereto with high potency to elicit IgE-secreting B-cell-bearing antibody-dependent cytotoxicity (ADCC) directed against the differentiation of B cells into IgE secretion)
FIG. 8 shows that various preparations of polyclonal anti-IgE EMPD antibodies produced using different IgE EMPD immunogen constructs (SEQ ID NOS: 88 to 93) are capable of expressing mIgE. Fc at an effector cell/target cell ratio of 30 L Ramos cells elicit ADCC. Polyclonal anti-IgE EMPD antibodies purified from guinea pig serum by protein A chromatography were used at a concentration of 10. Mu.g/mL. IL-2 stimulated mouse spleen cells were used as effector cells. Mouse anti-IgE monoclonal antibody 5D5 secretion was used as a positive control.
As shown in FIG. 8, all peptide immunogen constructs (SEQ ID NOS: 88-93) of long B cell epitope peptides having more than 20 amino acid residues elicit ADCC directed to the differentiation of B cells into IgE-secreting B cells bearing IgE, indicating that IgE serum levels can be reduced and inhibited upon immunization of a host with such peptide immunogen constructs.
vii) expansion of MHC coverage by use of IgE EMPD-derived peptide immunogen constructs with different promiscuous T helper cell epitopes
In designing pharmaceutical compositions for treating patients of different genetic backgrounds, it is important to allow the design to cover the largest population with different genetic backgrounds. Because promiscuous T helper cell epitopes derived from MVF or HBsAg represent one of the most potent T helper cell epitopes that provide such an increase in immunogenicity, combinations of peptide constructs containing both T helper cell epitopes can be designed to allow for synergistic immunogenic effects. It is expected that a mixture of two peptide immunogen constructs having the same B cell epitope may elicit a considerable immune response compared to the immune response elicited by the corresponding individual peptide constructs.
viii) epitope identification against fine specificity assays using immune serum (9 wpi) raised by various IgE EMPD peptide immunogen constructs
IgE-EMPD vaccines comprising IgE-EMPD peptide immunogen constructs are designed against a specific cyclic structure of C18-C39 located in the middle region of IgE-EMPD as a functional and structural target. This structure-based design aims at preserving the natural extracellular loop-like structure as an immunogenic target.
Four representative IgE-EMPD peptide fragments of 1-17 (SEQ ID NO: 7), 19-38 (SEQ ID NO: 8), 1-39 (SEQ ID NO: 5) and 7-40 (SEQ ID NO: 6) were used to design B cell epitope peptides which were attached to the B cell epitope peptide at either the amino or carboxy terminus(SEQ ID NO: 72) or +.>(SEQ ID NO: 73) to form a prototype peptide immunogen. An epsilon K linker or epsilon K-KKK (SEQ ID NO: 129) spacer was used between the B cells and Th epitopes to form the peptide immunogen constructs shown in Table 3 (SEQ ID NO:88 to 97). All peptide fragments with amino acids (aa) 1-39 and 7-40 were designed by cyclization to have a C18-C39 constrained cyclic structure.
Antibody reactivity against hyperimmune serum provided by guinea pigs immunized with their corresponding peptide immunogen constructs (SEQ ID NO:96, 97, 88, 89, 93) was assessed using ELISA assays coating microplates with individual IgE-EMPD B cell epitope peptides of 1-17 (SEQ ID NO: 7), 19-38 (SEQ ID NO: 8), 1-39 (SEQ ID NO: 5) and 7-40 (SEQ ID NO: 6). The results show the construct SEQ ID NO: 88. 89 and 93 raised high titers of antibodies against all four IGE EMPD B cell epitope peptides, whereas guinea pig antisera raised by IgE-EMPD 1-17 (SEQ ID NO: 96) and IgE-EMPD 19-38 (SEQ ID NO: 97) peptide immunogen constructs only had antibody reactivity with their corresponding B cell epitope peptides (e.g., SEQ ID NO:7 or 8), which were not cross-reactive with IgE EMPD B cell epitope peptides from non-corresponding adjacent epitopes (SEQ ID NO:8 or 7), indicating high specificity of immunogenicity, i.e., the designed immunogen constructs were able to elicit specific antibodies to react with IgE-EMPD corresponding B cell epitope domains (Table 5).
In a fine epitope identification assay to locate the binding site of an antibody to a specific residue within a region of interest, 50 overlapping 10-mer peptides (SEQ ID NOS: 10 to 58) were synthesized, which cover the 1 to 50 amino acid sequence regions from the first 8 amino acids of the CH4 carboxy terminus of IgE and IgE-EMPD. These 10-mer peptides were individually coated as solid phase immunoadsorbers onto 96 well microtiter plates. Pooled guinea pig antisera formulated in sample dilution buffer at a dilution ratio of 1:100 were added to microplate wells coated with 2.0 μg/mL of 10-mer peptide, followed by incubation at 37℃for 1 hour. After washing the microplate wells with wash buffer, horseradish peroxidase (HRP) -conjugated recombinant protein a/G was added and incubated for 30 minutes. After washing again with PBS, the substrate was added to the wells and absorbance at 450nm was measured using ELISA microplate analyzers to analyze the samples in a double format. Binding of immune serum elicited by IgE-EMPD peptide immunogens to corresponding IgE EMPD B cell epitope peptide-coated wells represents the greatest antibody binding signal.
Fine epitope identification showed that pooled guinea pig serum from IgE-EMPD 19-38 derived peptide immunogen constructs (SEQ ID NO:97, with non-circularized linear B cell epitopes, H19-R38) recognized the IgE EMPD 22-31 peptide (SEQ ID NO: 39) located in the IgE-EMPD middle region. It also reacted with IgE EMPD peptides 19-38 (SEQ ID NO: 8), 1-39 (SEQ ID NO: 5) and 7-40 (SEQ ID NO: 6) instead of 1-17 (SEQ ID NO: 7).
Antisera raised using IgE EMPD1-17 (SEQ ID NO:96, non-circularized linear B cell epitope, G1-L17) recognizes IgE EMPD 8-17 (SEQ ID NO: 25), and IgE EMPD1-17 (SEQ ID NO: 7), 1-39 (SEQ ID NO: 5) and 7-40 (SEQ ID NO: 6), but not 19-38 (SEQ ID NO: 8) peptides, located in the amino terminal region of IgE-EMPD. Antisera raised using IgE EMPD1-17 (SEQ ID NO: 96) and IgE EMPD 19-38 (SEQ ID NO: 97) immunogen constructs were not cross-reactive.
Interestingly, use is made ofTwo corresponding IgE-EMPD 1-39 immunogen constructs with epitopes linked to the amino-terminus (SEQ ID NO: 89) or the carboxy-terminus (SEQ ID NO: 88) of the IgE EMPD B-cell epitope peptide produced immune sera that recognized significantly different epitope binding patterns. The immune serum elicited by the immunogen construct (SEQ ID NO:89, UBTH1 at the amino terminus of the peptide) reacted strongly only with a 10-mer peptide (SEQ ID NO: 47) located aa 30-39 near the carboxy-terminal region of IgE-EMPD. It also reacted weakly with IgE EMPD 9-18 (SEQ ID NO: 26) located in the amino terminal region.
However, for other IgE-EMPD 1-39 immunogen constructs (SEQ ID NO:88, UBTH1 at the carboxy-terminus), the induced antisera reacted strongly with three discrete linear epitopes represented by IgE EMPD9-19 (SEQ ID NO:27 and 28), igE EMPD 19-31 (SEQ ID NO:37, 38, 39 and 40) and IgE EMPD 30-43 (SEQ ID NO:48, 49, 50, 51 and 52). The peptide immunogen construct (SEQ ID NO: 88) showed a stronger immunogenicity and a broader surface reaction than the IgE EMPD peptide immunogen construct (SEQ ID NO: 89). The much more potent immunogenicity associated with the peptide immunogen construct (SEQ ID NO: 88) was found to also show greater ADCC and apoptotic activity in IgE expressing lymphocytes than the IgE EMPD peptide immunogen construct (SEQ ID NO: 89).
In the case of the IgE-EMPD 7-40 immunogen construct (SEQ ID NO:93, having two UBTh 1's located at the amino-and carboxy-terminus of the IgE EMPD B cell epitope peptide), its induced antisera recognizes two major antigenic regions, which are similar to those recognized by IgE-EMPD1-39, as it can be identified by the peptide SEQ ID NO:35-41, igE EMPD 29-43 (SEQ ID NO: 45-51) in the IgE EMPD 18-33 region. The IgE-EMPD 7-40 immunogen construct (SEQ ID NO: 93) shares a similar epitope region as the IgE-EMPD1-39 immunogen construct (SEQ ID NO:88, UBTH1 is located at the carboxy terminus of the IgE EMPD B cell epitope peptide). IgE-EMPD1-39 (SEQ ID NO: 88) shows optimal efficacy in all designed peptide immunogen constructs, as shown by multiple functional assays related to the results shown in their fine epitope identification, as this immunogen construct elicits highly binding polyclonal antibodies that can recognize the broad surface covered by three or more B cell epitope peptides on the IgE-EMPD's extracellular membrane protein. The IgE EMPD30-43 (SEQ ID NO: 47-51) epitope region represents a very important B cell epitope region located in the carboxy-terminal region of the circular structure, adjacent to the basement membrane of the B cell carrying IgE, sensitive to antibody-mediated apoptosis and ADCC. Furthermore, the cyclized cyclic structure may present a better quality immunogen construct than its acyclic counterpart (table 5,SEQ ID NO:88vs SEQ ID NO:88 non-cyclized).
In summary, both the synthetic IgE-EMPD peptide immunogen constructs (IgE EMPD 1-39, SEQ ID NO: 88) and (IgE EMPD 7-40, SEQ ID NO: 93) were designed to have B cell epitope peptides represented by the cyclic structure within IgE EMPD linked to elicit a vigorous immune responseEpitope peptides, the polyclonal antibodies generated were directed against a novel epitope region (aa 29-43) located on the IgE-EMPD protein, which was close to the cell membrane because of the position at the carboxy-terminal region of the central loop structure, allowing as much cross-linking of the multiple cell membrane IgE as possible to induce ADCC and apoptosis to deplete IgE expressing B lymphocytes (table 5).
Example 7 concentrated antibody response believed to be directed against B cell epitopes of IgE EMPD peptide immunogen constructs only
It is well known that all carrier proteins used to boost immune responses against target B cell epitope peptides, such as Keyhole Limpet Hemocyanin (KLH) or other carrier proteins, such as Diphtheria Toxoid (DT) and Tetanus Toxoid (TT) proteins, can elicit more than 90% of antibodies against the boost carrier protein by chemically conjugating such B cell epitope peptides to the respective carrier proteins, while less than 10% of antibodies are against the target B cell epitope in the immune host.
It is therefore important to assess the specificity of antibodies raised by the IgE EMPD peptide immunogen constructs of the invention. Representative IgE EMPD peptide immunogen constructs (SEQ ID NO: 94) have a B cell epitope (SEQ ID NO: 5) linked to a heterologous T helper epitope by the spacer sequence εK-KKK (SEQ ID NO: 129)(SEQ ID NO: 72) was prepared for antigenicity evaluation. Will->(T helper cell epitope peptide for B cell epitope immunopotentiation) A microplate was coated and serum from immunized guinea pigs was used for testing and +.>Cross-reactivity of peptides. Table 7 shows that antibodies raised from IgE EMPD peptide immunogen constructs (SEQ ID NO: 94) are highly immunogenic against the corresponding target B cell epitope of IgE EMPD (SEQ ID NO: 5); however, it was found that most, if not all, of the immune serum pairs +.>The peptide (SEQ ID NO: 72) was not reactive.
In summary, the design of immunogens incorporating target B cell epitopes linked to carefully selected T helper cell epitopes results in a concentrated and pure immune response eliciting antibodies directed against only IgE EMPD B cell epitopes, not against Th epitopes to enhance the immune response. For pharmaceutical composition design, the more specific the immune response generated by the immunogen is, the more safe it provides for the composition. Thus, the IgE EMPD peptide immunogen constructs of the invention are highly effective in addition to being highly specific for their targets.
Example 8 Effect of immunotherapeutic allergy vaccine on the prophylactic and therapeutic model of serum IgE levels in genetically modified knock-in mice
Selecting SEQ ID NO:88 and SEQ ID NO:93 IgE EMPD peptide immunogen constructs IgE production in primary and secondary/memory responses was assessed in a proof of concept study using genetically engineered mice expressing human IgE (F1 offspring of huigex Balb/c knock-in mice).
Initially, igE EMPD peptide immunogens were prime-boost prior to sensitization with allergen (e.g., papain) challenge. Eight animals (n=8) were assigned to each of the six treatment groups and one placebo group for a total of 7 groups. For each of the two peptide immunogens, 3 subgroups were designed that utilized different doses of 9 (low), 18 (medium) and 40 (high) μg/dose in the i.m. route at weeks 0, 3 and 5. Peptide immunogens were formulated with ISA 51VG and CpG adjuvants to enhance immune responses. Mice in the placebo group were injected intramuscularly with vehicle only with formulation solution at weeks 0, 3 and 5. At weeks 10 and 16, all animal groups, including placebo group, were sensitized by the subcutaneous injection route with 50 μg papain and TiterMax Gold adjuvant (fig. 10).
The antibody response (IgG titers against the gamma 1-em67 fusion protein) of the two peptide immunogens was determined using ELISA assays as described in example 2. All mice developed strong antibody responses in six experimental groups immunized with different doses at weeks 0, 3, 5. ELISA data showed that serum samples from all experimental groups with high antibody titers could specifically bind to recombinant γ1-em67 fusion protein starting at week 3 and remained at high titers until week 20 (FIG. 11). In contrast, mice in the placebo group did not produce specific antibodies to the recombinant γ1-em67 fusion protein. These results indicate that all treatment groups can produce anti-IgE-EMPD antibodies with the potential to target IgE-expressing B lymphocytes, resulting in inhibition of IgE production. Figure 11 illustrates that high antibody titers lasted the entire 20 week trial period. These results also indicate that, even in the low dose group (9. Mu.g/dose) from each of the two vaccine immunogens, the peptide immunogens (S EQ ID NO:88 and SEQ ID NO: 93) were immunogenic to induce specific immune responses, resulting in high titers of anti-IgE EMPD antibodies.
The immune effect on IgE production in primary and memory responses of immunized mice was studied by measuring serum basal IgE levels and allergen-specific IgE levels using the analytical procedure described in example 2. Serum basal IgE levels before and after vaccination are shown in fig. 12, which demonstrates a gradual decline in serum basal IgE levels in mice in all treatment groups compared to serum basal IgE levels at the corresponding time points in placebo group. At week 10, the basal IgE levels were reduced to the minimum level in all six treatment groups prior to sensitization, while the basal serum IgE levels in the placebo group were not significantly altered. This result shows that basal IgE production was significantly inhibited in all treatment groups in animals receiving vaccine formulations containing IgE EMPD peptide immunogen constructs (SEQ ID NO:88 or 93) at week 10 compared to placebo groups. Figure 12 also shows that basal serum IgE levels in all treatment groups were inhibited throughout the 20 week trial period, even after two allergen sensitization at weeks 10 and 16.
Figures 13 and 14 show allergen-specific IgE production by first sensitization at week 12 and second sensitization at week 18 with papain, respectively. These results indicate that the two peptide immunogen constructs (SEQ ID NOS: 88 and 93) significantly inhibited papain-specific IgE levels after the first and second allergen sensitization compared to placebo. No substantial difference between the three dose levels was observed in either of the two peptide immunogens. As shown in FIG. 13, after allergen-specific IgE was produced in the primary reaction at week 12, the peptide immunogen (SEQ ID NO: 88) was expressed slightly better than (SEQ ID NO: 93) in this study. The two IgE EMPD peptide immunogen constructs exhibited significant inhibition of allergen-specific IgE production in both the primary and memory responses compared to placebo group (fig. 13 and 14).
To further investigate the potential therapeutic effects of IgE-EMPD peptide immunogen constructs (SEQ ID NO:88 and SEQ ID NO: 93) targeting IgE-expressing B cells to inhibit IgE production and treat IgE-mediated allergic diseases, additional experimental protocols were designed to assess the effect of these two peptide immunogen constructs on sensitization/recall (recovery) responses in huige knockout mice. Six animals were assigned to each of the two treatment groups (n=6), while four animals were used for the placebo group (n=4), for a total of three groups. All groups of mice received two sensitization, before and after peptide vaccination, respectively, by subcutaneous route at week 0 and footpad route at week 12, using 50 μg papain/TiterMax Gold adjuvant. The prime-boost regimen was evaluated in two treatment groups, with a dose of 40 μg/0.1mL of the dosage form containing one of the two peptide immunogen constructs at weeks 3, 6 and 8, while the placebo group was administered with adjuvant vehicle alone (see figure 15).
The results show that all groups, including placebo groups, were sensitized with papain, that high titers of papain-specific IgGs were elicited in all three groups after week 2, and remained high until week 6 (last period of observation). Total serum IgE and papain-specific IgE levels reached maximum at week 2, then began to decline gradually at week 3 and returned to baseline at week 6 (fig. 16).
Vaccination with IgE EMPD peptide immunogen constructs elicits high titers of antibodies that specifically recognize the γ1-em67 fusion protein. After three peptide immunogen injections at weeks 3, 6 and 8, the elicited antibody IgG titers (against anti- γ1-em67 or anti-IgE-EMPD) steadily increased and reached the highest level at week 10 (data not shown). At week 12, neither treatment group showed elevated papain-specific IgE levels after the second papain sensitization; however, papain-specific IgE levels increased significantly in the group of Yu Anwei at weeks 12 and 13, and then decreased to lower levels at week 14 (data not shown).
The study results showed that, compared to placebo group, SEQ ID NO:88 and 93 are capable of eliciting specific humoral immune responses in organisms to prevent memory B cell recall proliferation responses that completely block papain-specific IgE production by papain recall (recall) at week 12 (fig. 17). Overall, this study indicated that the antibody response induced by the IgE EMPD peptide immunogen constructs of the present disclosure not only inhibited allergen-specific serum IgE production from primary sensitization, but also stabilized allergen-specific serum IgE from recall of secondary allergen challenge. This study result demonstrates that the disclosed invention provides potentially effective therapeutic vaccines for the treatment of IgE-mediated allergic diseases (e.g., asthma). After careful study of the efficacy to attenuate allergen-specific IgE, the two peptide immunogens (SEQ ID NO:88 and SEQ ID NO: 93) exhibited similar efficacy in inhibiting allergen-specific IgE production.
EXAMPLE 9 dose and formulation studies in cynomolgus monkeys by immunogenicity assessment of prototype immunotherapeutic allergy vaccine formulations
a. The whole object is as follows:
the purpose of this study was to evaluate the use of the selected IgE EMPD peptide immunogen construct SEQ ID NO:88 effects of intramuscular immunization on immunogenicity. Before conducting human trials, cynomolgus monkeys were selected as animal models for assessing the immunogenicity of prototype IgE-EMPD peptide vaccine formulations and dosing regimens. The representative peptide immunogen construct (SEQ ID NO: 88) was formulated into two common dosage forms. In the first dosage form, the composition is prepared with Montanide TM Prior to mixing ISA51 to form a water-in-oil emulsion, SEQ ID NO:88 IgE EMPD peptide immunogen constructs were made into stabilized immunostimulatory complexes with CpG (part a study). In the second formulation, prior to forming a suspension dosage form with ADJUPHOS, SEQ ID NO:88 with CpG to form a stabilized immunostimulatory complex (part B). In this comprehensive immunogenicity study, four doses of from 30 μg, 100 μg, 300 μg to 1000 μg per dose were evaluated in each dosage form.
b. Planning the abstract of a book
2.5-4.0kg adult cynomolgus monkeys were selected to assess the effect of IgE EMPD peptide immunogens on immunogenicity and serum cynomolgus IgE levels. A total of 20 cynomolgus monkeys were divided into 10 groups: placebo-controlled animals (n=2) were treated with adjuvant alone (Montanide TM ISA51 plus CpG oligodeoxynucleotide) or (ADJUPHOS plus CpG oligodeoxynucleotide)) And (3) injection. The experimental animals were injected with IgE EMPD peptide immunogen (SEQ ID NO: 88) at doses of 30, 100, 300 and 1,000 μg per group (total 500 μl vaccine volume per animal; n=2 per group, male 1 and female 1). A total of three intramuscular immunizations were administered at weeks 0, 3 and 6. All cynomolgus monkeys were monitored for immunogenicity and serum IgE levels at weeks 0, 3, 6, 8, 10, 12, 14, 16, 20 and 24.
c. Determination of anti-IgE EMPD antibody titers
All animals were bled at weeks 0, 3, 6, 8, 10, 12, 14, 16, 20 and 24. Serum from each blood collection was isolated to determine anti-IgE EMPD antibody titers using gamma 1-cynoem 67 ELISA. Placebo-treated animals had few detectable anti-IgE EMPD antibody titers (fig. 18A and 18B). However, all animals receiving three immunizations had detectable IgG antibody titers against IgE EMPD B cell epitopes, which had peak titers at weeks 8 to 12. This specific reactivity was maintained in a dose-dependent manner throughout the 24 week study period (fig. 18A and 18B). All animals were found to have high antisera titers against gamma 1-cynoem 67 recombinant protein, with a dose of 300 μg per dose in 0.5mL being optimal for both dosage forms, since a 1,000 μg dose formulation would result in an excess of peptide immunogen in the respective formulation relative to ISA51V or ADJUPHOS adjuvant. The results show that formulations containing ISA51/CpG ODN have higher immunogenic results than formulations using ADJUPHOS/CpG ODN. The antibody response against IgE EMPD B-cell epitopes was enhanced with each peptide immunogen boost, then the anti-IgE EMPD titers decreased gradually over time. Significantly higher antisera titers continued throughout the 24 week study (fig. 18A and 18B).
Example 10 demonstration of the efficacy of alternative immunotherapeutic allergy vaccine in cynomolgus monkey
a. The whole object is as follows:
the purpose of this study was to assess the effect of intramuscular immunization with IgE EMPD peptide immunogens (where this sequence is derived from autologous mIgE) on immunogenicity and serum IgE levels in cynomolgus monkeys, an animal model that closely mimics human IgE production, over a 20 week period. IgE EMPD of mIgE is evolutionarily conserved in non-human primates (e.g., monkeys and apes in the new and old world), and such IgE EMPD counterpart sequences are not found in other species (e.g., rodents, rabbits, and canines). The amino acid sequence of cynomolgus monkey IgE EMPD (SEQ ID NO: 127) has a high degree of sequence identity (90%) with the amino acid sequence of human IgE EMPD (SEQ ID NO: 2).
b. Planning the abstract of a book
2.5-4.0kg adult cynomolgus monkeys were selected to assess the effect of IgE EMPD peptide immunogens on immunogenicity and serum cynomolgus IgE levels. A total of 12 cynomolgus monkeys were divided into 3 groups: placebo-controlled animals (n=4, 2 males and 2 females) were treated with adjuvant only (Montanide TM ISA 51 plus CpG oligodeoxynucleotide); experimental animals (total 500 μl vaccine volume per animal; n=4, 2 males and 2 females per group) were injected with IgE EMPD peptide immunogen (SEQ ID NO:125 or 126) at a dose of 300 μg. A total of three intramuscular immunizations were administered at weeks 0, 3 and 6. All cynomolgus monkeys were monitored for immunogenicity and serum IgE levels at weeks 0, 3, 6, 8, 10, 12, 14, 16, 18 and 20.
c. Determination of anti-IgE EMPD antibody titers
All animals were bled at weeks 0, 3, 6, 8, 10, 12, 14, 16, 18 and 20. Serum from each blood collection was isolated to determine anti-IgE EMPD antibody titers using gamma 1-cynoem 67 ELISA. Placebo-treated animals had no detectable anti-IgE EMPD antibody titers (fig. 19). However, it is accepted to use SEQ ID NO:125 or SEQ ID NO: all animals immunized 126 three times had detectable IgG antibody titers against IgE EMPD B cell epitopes, which had peak titers at weeks 8 to 12 (fig. 19). This specific reactivity was maintained for the entire 20 week study period. In addition, all immunized animals produced specific IgM and IgA antibody titers throughout the 20 week period (fig. 20).
d. Determination of serum IgE levels
All animals were bled at weeks 0, 3, 6, 8, 10, 12, 14, 16, 18 and 20. Serum from each blood collection was isolated to determine serum IgE using a quantitative cynomolgus IgE ELISA. Basal IgE levels of placebo group varied during the survival period. In administering SEQ ID NO: the IgE reduction observed in the 125 group was statistically significant, whereas upon administration of SEQ ID NO: also shown in the group 126 is the trend of IgE reduction during monitoring (fig. 19).
e. Results
The effect of IgE EMPD peptide immunogens on immunogenicity against white body mIgE and serum IgE concentration in adult cynomolgus monkey serum samples in surrogate models was evaluated. In this proof of concept study, cpG ODN and Montanide were utilized TM Four animals in each group were dosed as placebo control with ISA 51 mixture and 300 μg cynomolgus IgE EMPD peptide immunogen with SEQ ID NO:125 or 126, forming a proprietary immunostimulatory complex (ISC) with CpG oligodeoxynucleotide (CpG ODN) and with Montanide TM ISA 51 adjuvant formulation, 8 adult cynomolgus monkeys (n=4 per group) were immunized at weeks 0, 3 and 6. SEQ ID NO:125 and 126 are human IgE EMPD immunogens SEQ ID NO:93 and 88. With CpG ODN/Montanide TM Two cynomolgus derived immunogens formulated in ISA 51 produced a strong anti-IgE EMPD IgG antibody response in all animals (fig. 19). In addition, all animals produced IgM and IgA antibodies against IgE EMPD (fig. 20). A downward trend in basal cynomolgus serum IgE was also observed (fig. 21). No adverse injection site reactions were recorded. This study demonstrated enhanced performanceSynthetic IgE EMPD peptide immunogens (SEQ ID NOS: 125 and 126), the sequences of which are derived from autologous, of T cell epitopes are capable of eliciting strong anti-IgE EMPD antibody responses, leading to inhibition of IgE production and a trend towards reduced serum IgE levels in basal cynomolgus monkeys.
f. Conclusion(s)
Cynomolgus monkeys were injected 3 times by intramuscular route over a 20 week period using IgE EMPD peptide immunogen (SEQ ID NOs: 125 and 126) or placebo control. Animals have good overall tolerance and develop immune tolerance. All immunized cynomolgus monkeys produced transient specific IgM antibodies, with simultaneous needle productionEfficient and sustained IgG (up to 10) 5 ) And IgA (up to 10) 4 ) Antibody titers. A decrease in basal IgE levels was observed in each and all of the responders. These results support the mode of action of anti-IgE-EMPD antibodies whereby the antibodies target B cells expressing membrane-bound IgE resulting in subsequent inhibition of IgE production.
TABLE 1
IgE for serological analysis -EMPD Peptides and fragments thereof
Table 1 (subsequent)
TABLE 2
In IgE- EMPD Selected promiscuous T helper cell epitopes for use in the design of derived peptide immunogen constructs
TABLE 3 Table 3
In IgE- EMPD IgE using pathogen protein derived Th epitopes (including ideal artificial Th epitopes) for eliciting specific antibodies in peptide immunogen construct design -EMPD Antigenic enhancement of peptide fragments
Table 3 (Xuang)
Peptides are cyclized by cysteine disulfide bonds, with the bottom line below the cysteines.
TABLE 4 Table 4
IgE -EMPD Immunogenicity evaluation of derived peptide immunogen constructs in guinea pigs
a Groups 1-6: immunization was received at 0, 2, 4, 6, 8 and 10 wpi: blood was collected at 0, 4, 6, 8, 10, 12 and 14 wpi.
/>
/>
/>
/>
/>

Claims (4)

1. An IgE EMPD peptide immunogen construct, wherein the peptide immunogen construct is selected from the group consisting of SEQ ID NOs 88-90, 94, 95, 98-124 and 130.
2. A composition comprising the peptide immunogen construct of claim 1.
3. A pharmaceutical composition comprising:
a. the peptide immunogen construct of claim 1; and
b. pharmaceutically acceptable delivery vehicles and/or adjuvants.
4. A pharmaceutical composition according to claim 3, wherein
The IgE EMPD peptide immunogen construct is mixed with CpG Oligodeoxynucleotides (ODNs) to form a stabilized immunostimulatory complex.
CN201780098083.4A 2017-12-31 2017-12-31 Peptide immunogens targeting membrane-bound IgE for the treatment of IgE-mediated allergic diseases and dosage forms thereof Active CN111565801B (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US2017/069174 WO2019133024A1 (en) 2017-12-31 2017-12-31 Peptide immunogens and formulations thereof targeting membrane-bound ige for treatment of ige mediated allergic diseases

Publications (2)

Publication Number Publication Date
CN111565801A CN111565801A (en) 2020-08-21
CN111565801B true CN111565801B (en) 2023-11-10

Family

ID=67068075

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201780098083.4A Active CN111565801B (en) 2017-12-31 2017-12-31 Peptide immunogens targeting membrane-bound IgE for the treatment of IgE-mediated allergic diseases and dosage forms thereof

Country Status (13)

Country Link
US (1) US20230270848A1 (en)
EP (1) EP3731934A1 (en)
JP (1) JP7239203B2 (en)
KR (1) KR20200115522A (en)
CN (1) CN111565801B (en)
AU (1) AU2017445171A1 (en)
BR (1) BR112020013352A2 (en)
CA (1) CA3087036A1 (en)
MX (1) MX2020006937A (en)
RU (1) RU2769983C2 (en)
SG (1) SG11202006215TA (en)
TW (1) TWI804553B (en)
WO (1) WO2019133024A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA3168991A1 (en) * 2020-01-23 2021-07-29 United Biomedical, Inc. Peptide immunogens targeting pituitary adenylate cyclase-activating peptide (pacap) and formulations thereof for prevention and treatment of migraine

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1167491A (en) * 1994-10-25 1997-12-10 美国联合生物医学公司 Synthetic IgE membrane anchor peptide immunogens for the treatment of allergy
US9408897B2 (en) * 2002-06-20 2016-08-09 The Trustees Of The University Of Pennsylvania Vaccines for suppressing IgE-mediated allergic disease and methods for using the same

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI227241B (en) * 1998-06-20 2005-02-01 United Biomedical Inc IgE-CH3 domain antigen peptide, peptide conjugate containing the same, and pharmaceutical composition for treating allergies containing the peptide conjugate
US20040156838A1 (en) 2000-09-06 2004-08-12 Steen Klysner Method for down-regulating ige
US6906169B2 (en) * 2001-05-25 2005-06-14 United Biomedical, Inc. Immunogenic peptide composition comprising measles virus Fprotein Thelper cell epitope (MUFThl-16) and N-terminus of β-amyloid peptide
CN102482351B (en) * 2009-02-25 2015-03-18 中央研究院 Anti-c[epsilon]mx antibodies capable of binding to human mIgE on b lymphocytes
US9102752B2 (en) * 2013-03-15 2015-08-11 United Biomedical, Inc. Peptide vaccine for prevention and immunotherapy of dementia of the Alzheimer's type

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1167491A (en) * 1994-10-25 1997-12-10 美国联合生物医学公司 Synthetic IgE membrane anchor peptide immunogens for the treatment of allergy
US9408897B2 (en) * 2002-06-20 2016-08-09 The Trustees Of The University Of Pennsylvania Vaccines for suppressing IgE-mediated allergic disease and methods for using the same

Also Published As

Publication number Publication date
CN111565801A (en) 2020-08-21
JP7239203B2 (en) 2023-03-14
RU2020125383A3 (en) 2022-01-31
TW201930346A (en) 2019-08-01
CA3087036A1 (en) 2019-07-04
US20230270848A1 (en) 2023-08-31
EP3731934A1 (en) 2020-11-04
SG11202006215TA (en) 2020-07-29
WO2019133024A1 (en) 2019-07-04
JP2021515744A (en) 2021-06-24
RU2769983C2 (en) 2022-04-12
RU2020125383A (en) 2022-01-31
KR20200115522A (en) 2020-10-07
MX2020006937A (en) 2020-10-05
TWI804553B (en) 2023-06-11
AU2017445171A1 (en) 2020-08-13
BR112020013352A2 (en) 2020-12-01

Similar Documents

Publication Publication Date Title
AU2019246916A1 (en) Peptide vaccine for prevention and immunotherapy of dementia of the Alzheimer&#39;s type
JP6466327B2 (en) Immunomodulating vaccine
AU2018383708B2 (en) Peptide immunogens of IL-31 and formulations thereof for the treatment and/or prevention of atopic dermatitis
CA2912736C (en) Gastrin peptide immunogenic composition
TW202039587A (en) Artificial promiscuous t helper cell epitopes as immune stimulators for synthetic peptide immunogens
JP2022515934A (en) Peptide immunogens targeting calcitonin gene-related peptides (CGRP) and their formulations for the treatment and prevention of migraine
CN111565801B (en) Peptide immunogens targeting membrane-bound IgE for the treatment of IgE-mediated allergic diseases and dosage forms thereof
TW202142551A (en) Peptide immunogens targeting pcsk9 and formulations thereof for prevention and treatment of pcsk9-mediated disorders
TWI823051B (en) Peptide immunogens targeting pituitary adenylate cyclase-activating peptide (pacap) and formulations thereof for prevention and treatment of migraine
JP2023519104A (en) Peptide immunogens targeting IAPP for preventing and treating disorders associated with aggregated islet amyloid polypeptide (IAPP)

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant